Download Evidence-based clinical practice guidelines for the use of

Document related concepts

Patient safety wikipedia , lookup

Adherence (medicine) wikipedia , lookup

Management of multiple sclerosis wikipedia , lookup

Transcript
Evidence-based clinical practice
guidelines for the use of recombinant
and plasma-derived FVIII and FIX
products
Released June 2006
Disclaimer:
The recommendations in this document are designed to provide information to
assist decision-making and have been developed based upon the best available
published evidence as at October 2004 to the parties involved in developing the
Guidelines [ie, the Jurisdictional Blood Committee (JBC) FVIII & FIX Working
Party, the Commonwealth of Australia, the Australian Haemophilia Centres
Directors Organization (AHCDO) and Adelaide Health Technology Associates
(AHTA)].
However, this document is not a substitute for individual medical advice
obtained from the attending clinician. The recommendations in this document
should be used subject to the clinician's judgement and the patient's preference
in each individual case, with the attending clinician having ultimate responsibility
for the appropriate choice of treatment. The recommendations also assume that
a correct diagnosis has been made and that the severity of the relevant disorder
has been properly ascertained.
Accordingly, each of the parties involved in developing the Guidelines and their
employees expressly disclaims, and accepts no responsibility for, any
consequences arising from relying upon the information contained in this
document.
It is recommended these guidelines be reviewed between 2008 and 2010. After
this time, these Guidelines should not be used without first determining their
currency. At the time of publication, this can be done by checking the NBA
website at www.nba.gov.au or the AHCDO website at www.ahcdo.org.au.
ISBN: 0 – 9775298 - 0 - 0
© Commonwealth of Australia 2006
This work is copyright. Apart from any use as permitted under the Copyright Act 1968,
no part may be reproduced by any process without prior written permission from the
Commonwealth. Requests and inquiries concerning reproduction and rights should be
addressed to the Commonwealth Copyright Administration, Attorney General’s
Department, Robert Garran Offices, National Circuit, Barton ACT 2600 or posted at
http://www.ag.gov.au/cca
Use of recombinant and plasma-derived Factor VIII and IX
This systematic literature review and clinical practice guidelines document was
developed for the Jurisdictional Blood Committee (JBC) FVIII & FIX Working
Party, which was established to oversee the development process.
Reviewers
Petra Bywood BSc (Hons), PhD
Research Officer, Adelaide Health Technology Assessment, Department of Public Health,
University of Adelaide, Adelaide, SA 5005
Skye Newton B Psych (Hons)
Research Officer, Adelaide Health Technology Assessment, Department of Public Health,
University of Adelaide, Adelaide, SA 5005
Linda Mundy BSc, DPH
Research Officer, Adelaide Health Technology Assessment, Department of Public Health,
University of Adelaide, Adelaide, SA 5005
Tracy Merlin BA (Hons), MPH
Manager, Adelaide Health Technology Assessment, Department of Public Health, University
of Adelaide, Adelaide, SA 5005
Ben Saxon MBBS, FRACP, FRCPA
Transfusion Medicine Specialist, Australian Red Cross Blood Service-Discovery, Department of
Haematology, Women’s and Children’s Hospital, North Adelaide, SA 5006
Janet Hiller MPH, PhD
Professor of Public Health and Director, Adelaide Health Technology Assessment, Department
of Public Health, University of Adelaide, Adelaide, SA 5005
i
Table of Contents
Summary of Guideline Recommendations.............................................................. 2
Introduction ..................................................................................................................... 2
Haemophilia A, without inhibitors ............................................................................... 5
Haemophilia A, with inhibitors ..................................................................................... 7
Haemophilia B, without inhibitors.............................................................................. 10
Haemophilia B, with inhibitors.................................................................................... 12
von Willebrand disease ................................................................................................. 13
Other rare bleeding disorders ...................................................................................... 15
Acquired haemophilia ................................................................................................... 16
Infant delivery ................................................................................................................ 18
Background.............................................................................................................21
Normal haemostasis ...................................................................................................... 21
Haemophilia ................................................................................................................... 21
von Willebrand disease ................................................................................................. 23
Other rare bleeding disorders ...................................................................................... 24
Complications of treatment ......................................................................................... 25
Approach to Guideline Development .................................................................... 27
How do we develop clinical practice guidelines? ...................................................... 27
Research questions ........................................................................................................ 28
Overview of methodology ........................................................................................... 28
Selection of Products ..............................................................................................31
Adjunctive haemostatic agents .............................................................................. 37
Treatment of Bleeding Episodes ............................................................................51
Haemophilia A, without inhibitors ............................................................................. 51
Haemophilia B, without inhibitors.............................................................................. 65
von Willebrand disease ................................................................................................. 69
Other rare bleeding disorders ...................................................................................... 76
Prophylaxis............................................................................................................. 77
Haemophilia ................................................................................................................... 78
von Willebrand disease ................................................................................................. 85
Other rare bleeding disorders ...................................................................................... 86
Inhibitors and Tolerisation .................................................................................... 87
Treatment of bleeding episodes in patients with inhibitors .................................... 90
Tolerisation protocols ................................................................................................... 93
Treatment of patients with acquired haemophilia .................................................... 98
Surgical and Dental Procedures ........................................................................... 105
Use of recombinant and plasma-derived Factor VIII and IX
ii
Haemophilia A and B.................................................................................................. 105
von Willebrand disease ............................................................................................... 110
Delivery of Infants................................................................................................. 118
Haemophilia ................................................................................................................. 121
von Willebrand disease ............................................................................................... 125
Other Aspects of Management ............................................................................. 131
Comprehensive care .................................................................................................... 131
Management of bleeding episodes ............................................................................ 132
Issues in treating young patients................................................................................ 132
Summary and Conclusions ................................................................................... 134
Areas for further research .......................................................................................... 140
Glossary and Abbreviations .................................................................................. 143
References ............................................................................................................. 147
Appendix A Terms of Reference for Undertaking Guidelines .......................... 160
A. Services..................................................................................................................... 160
B. Required contract material .................................................................................... 160
Appendix B Working Party Membership .......................................................... 161
Contractors ................................................................................................................... 161
National Blood Authority........................................................................................... 161
Recombinant FVIII and FIX Working Party Members ........................................ 162
Appendix C
Australian haemophilia treatment centres ................................... 163
Appendix D Currently available product information....................................... 164
Haemophilia A (without inhibitors).......................................................................... 164
Haemophilia A (with inhibitors)................................................................................ 168
Haemophilia B (without inhibitors) .......................................................................... 171
Haemophilia B (with inhibitors)................................................................................ 174
von Willebrand disease ............................................................................................... 176
Appendix E
Methodology.................................................................................. 178
Appendix F Specialty Websites.......................................................................... 184
Haemophilia websites ................................................................................................. 184
Other relevant medical and health websites ............................................................ 186
Appendix G Consensus Clinical Practice Guidelines........................................ 187
Previous Australian clinical guidelines on treatment and prophylaxis................. 187
Clinical guidelines on prophylactic treatment using factors VIII and IX ........... 190
Guidelines for the treatment of inhibitors in haemophilia A................................ 191
Protocols for dental procedures in patients with haemophilia A, B or von
Willebrand disease ...................................................................................................... 196
iii
Use of recombinant and plasma-derived Factor VIII and IX
Appendix H Critical Appraisal Checklists......................................................... 197
Systematic review critical appraisal checklist ........................................................... 197
Checklist for appraising the quality of intervention studies .................................. 198
Checklist for the critical appraisal of case series ..................................................... 202
Checklist for assessing the clinical importance of benefit or harm...................... 203
Checklist for assessing the relevance of outcomes ................................................. 204
Appendix I Evidence Tables..............................................................................205
Summary tables for intervention studies.................................................................. 205
Study profiles of included studies ............................................................................. 230
Use of recombinant and plasma-derived Factor VIII and IX
iv
List of Tables
Table 1.
Classification of haemophilia ................................................................................ 22
Table 2.
Evidence dimensions.............................................................................................. 29
Table 3.
Designations of levels of evidence ....................................................................... 30
Table 4.
Fresh plasma products not virally inactivated .................................................... 31
Table 5.
Plasma-derived coagulation factor concentrates ................................................ 32
Table 6.
Recombinant coagulation factor concentrates ................................................... 33
Table 7.
Prothrombin complex concentrates (PCCs)....................................................... 34
Table 8.
Coagulation factor concentrates for less common disorders ........................... 35
Table 9.
Adjunctive haemostatic agents.............................................................................. 37
Table 10. Effectiveness of DDAVP for haemostatic control ........................................... 40
Table 11. Adverse events associated with use of DDAVP................................................ 44
Table 12. Effectiveness of tranexamic acid .......................................................................... 48
Table 13. Safety of tranexamic acid ....................................................................................... 49
Table 14. Safety of fibrin glue ................................................................................................ 50
Table 15. FVIII recovery following treatment with rFVIII in haemophilia A
patients ..................................................................................................................... 53
Table 16. Number of infusions required to control haemostasis in haemophilia
A patients ................................................................................................................. 55
Table 17. Development of inhibitors in haemophilia A patients treated with
recombinant factor VIII ........................................................................................ 59
Table 18. Exposure days prior to inhibitor development in haemophilia A
patients treated with recombinant factor VIII ................................................... 62
Table 19. Adverse events in haemophilia A patients treated with recombinant
factor VIII................................................................................................................ 63
Table 20. FIX recovery following treatment of bleeding episodes in
haemophilia B patients........................................................................................... 66
Table 21. Number of FIX infusions required to control haemostasis in
haemophilia B patients........................................................................................... 67
Table 22. Development of inhibitors in haemophilia B patients treated with
recombinant factor IX ........................................................................................... 67
Table 23. Exposure days prior to inhibitor development in haemophilia B
patients treated with recombinant factor IX....................................................... 68
Table 24. Adverse events in haemophilia B patients treated with recombinant
factor IX................................................................................................................... 68
Table 25. Number of patients listed on Australian Bleeding Disorders Registry
for vWD subtypes................................................................................................... 69
Table 26. Methods of testing efficacy of treatments for von Willebrand disease........... 70
v
Use of recombinant and plasma-derived Factor VIII and IX
Table 27. Effectiveness of plasma-derived factor VIII/von Willebrand factor
concentrates in controlling bleeding episodes .................................................... 73
Table 28. Safety of plasma-derived products for von Willebrand disease for
treating bleeding episodes...................................................................................... 75
Table 29. Pettersson radiological score of haemophilic arthropathy................................ 77
Table 30. Factor recovery following prophylactic treatment with recombinant
factors in haemophilia A or B patients ................................................................ 81
Table 31. Inhibitor development in young children undergoing prophylaxis................. 83
Table 32. Adverse events in haemophilia A or B patients undergoing
prophylactic treatment with recombinant factors.............................................. 83
Table 33. Effectiveness of prophylaxis in von Willebrand disease................................... 86
Table 34. Products used for haemophilia patients with inhibitors ................................... 89
Table 35. Summary of efficacy of interventions to control bleeding in
haemophilia A patients with inhibitors................................................................ 92
Table 36. Adverse events associated with immunosuppressant therapy........................ 102
Table 37. Management of patients undergoing surgical procedures .............................. 105
Table 38. Haemostatic control in haemophilia patients undergoing surgery ................ 107
Table 39. Haemostatic control in haemophilia patients undergoing dental
procedures.............................................................................................................. 108
Table 40. Development of inhibitors in haemophilia patients undergoing
surgical procedures ............................................................................................... 109
Table 41. Adverse events in haemophilia patients undergoing surgical or dental
procedures.............................................................................................................. 110
Table 42. Effectiveness of adjunctive haemostatic agents in surgical and dental
procedures for patients with von Willebrand disease...................................... 111
Table 43. Effectiveness of local therapies, local therapies with DDAVP, and
local therapies with factor concentrates for dental surgery in von
Willebrand disease patients.................................................................................. 112
Table 44. Effectiveness of plasma-derived factor VIII/von Willebrand factor
concentrates for surgery or dental procedures in von Willebrand
patients ................................................................................................................... 113
Table 45
Adverse events associated with the use of plasma-derived factor
VIII/vWF concentrates....................................................................................... 116
Table 46. Antenatal diagnosis of haemophilia ................................................................... 119
Table 47. Effectiveness of antenatal diagnosis .................................................................. 120
Table 48. Safety of antenatal diagnosis ............................................................................... 121
Table 49. Infant delivery and haemophilia ......................................................................... 124
Table 50. Infant delivery and von Willebrand disease ...................................................... 127
Table 51. A matrix of treatment recommendations for patients with
haemophilia A (without inhibitors to FVIII).................................................... 164
Use of recombinant and plasma-derived Factor VIII and IX
vi
Table 52. A matrix of treatment recommendations for patients with
haemophilia A (with inhibitors to FVIII).......................................................... 168
Table 53. A matrix of treatment recommendations for patients with
haemophilia B (without inhibitors to FIX) ....................................................... 171
Table 54. A matrix of treatment recommendations for patients with
haemophilia B (with inhibitors to FIX)............................................................. 174
Table 55. A matrix of treatment recommendations for patients with von
Willebrand disease ................................................................................................ 176
Table 56. Bibliographic databases........................................................................................ 179
Table 57. Other sources of evidence (1966–9/2004) ....................................................... 180
Table 58. Search terms utilised............................................................................................. 181
Table 59. Number of citations initially retrieved and then retained at each phase....... 182
vii
Use of recombinant and plasma-derived Factor VIII and IX
Foreword
From an historical perspective, these guidelines originated in October 2003 when the
Australian Health Ministers’ Conference (AHMC) agreed to increase access to
recombinant clotting factors and to continued access to plasma-derived products where
appropriate for all Australians with haemophilia, and subject to funding. In 2004, all
Australian Governments announced their agreement to fund the increased access
commencing 1 July 2004, conditional on the development of appropriate clinical practice
guidelines and tolerisation protocols.
Australian
consensus
Directors’
guidelines
situation.
guidelines for treating people with haemophilia at this stage were still
based, and modified from the United Kingdom Haemophilia Centre
Organisation (UKHCDO) Executive Committee Guidelines. These new
however, are evidence-based and specifically developed for the Australian
On 5 December 2003, The Jurisdictional Blood Committee (JBC) met and agreed to the
development, Terms of Reference and membership of a Working Party to oversee the
development of these new guidelines. The Working Party was established in January
2004, and consisted of members from the Australian Haemophilia Centre Directors
Organization (AHCDO) for clinical input, the Haemophilia Foundation of Australia
(HFA) for consumer representation, and the JBC. The NBA provided secretariat support
to the Working Party.
On 18 August 2004, Adelaide Health Technology Assessment (AHTA) was awarded the
contract to develop the Guidelines. Their methodology a-priori was evidence-based, and
evolved from a comprehensive systematic literature review that integrated evidence from
empirical research using a systematic and unbiased approach.
Recommendations outlined in these guidelines are based on the highest level of evidence
available. In the absence of good quality comparative studies, many recommendations are
based on lower levels of evidence such as case series. Where empirical evidence was
unavailable, expert opinion relevant to Australia has been included and stated
accordingly. The level of evidence is indicated for each recommendation in this
document.
A list of reviewers for the literature review is included inside the front cover of this
document.
A list of Contractors, JBC FVIII & FIX Working Party members and NBA support staff
is available at page 161 of this document.
The content of these guidelines was approved by the JBC on 11 March 2005, and by
AHMAC in June 2006. The guidelines were endorsed by AHCDO on 15 July 2005.
These guidelines can be used by both clinicians and their patients to guide treatment of
haemophilia in Australia, and are based on the most up-to-date evidence available at the
time of development. However, the ultimate responsibility for the care and treatment of
patients with haemophilia lies with the attending doctor.
Use of recombinant and plasma-derived Factor VIII and IX
1
Summary of Guideline Recommendations
Introduction
Rationale for developing clinical practice guidelines
The previous Australian clinical guidelines were modified from the United Kingdom
Haemophilia Centre Directors Organisation (UKHCDO) Executive Committee
guidelines by the Haemophilia Foundation Australia Medical Advisory Panel in March
2000 (Haemophilia Foundation Australia Medical Advisory Panel 2000). These guidelines
were based on a non-systematic review of published evidence and were largely
consensus-based.
The Australian Health Ministers’ Advisory Council (AHMAC) Blood and Blood
Products Committee Report of the Working Party on the Supply and Use of Factor VIII and
Factor IX in Australia was published in April 2003. This report made several
recommendations about the extended use of recombinant factors VIII and IX. These
recommendations were endorsed in the Report of the Expert Advisory Group on Hepatitis C
and Plasma in 1990 (Barraclough Report) tabled in the Senate in May 2003 and later in
The Senate Community Affairs References Committee report Hepatitis C and the blood
supply in Australia in June 2004.
In response to the AHMAC Working Party report, the National Blood Authority was
requested by the Jurisdictional Blood Committee (JBC) in December 2003 to
commission the development of evidence-based clinical practice guidelines and national
tolerisation protocols for the use of recombinant and plasma-derived factor VIII and IX
products.
On 30 August 2004, the Australian government announced funding for access to
recombinant factors VIII and IX for haemophilia patients (Department of Health and
Ageing 2004). The Jurisdictional Blood Committee (JBC) indicated that the new policy
would be implemented throughout Australia soon after 1 October 2004. Accordingly,
access to recombinant factor VIII and IX has been extended to people who were
ineligible under the previous policy, and these evidence-based guidelines refer primarily
to the use of recombinant products.
Approach to developing evidence-based clinical practice guidelines
These evidence-based clinical practice guidelines evolved from a systematic literature
review that integrated evidence from empirical research using a systematic, unbiased
approach. Recommendations outlined in these guidelines are based on the highest level
of evidence available. In the absence of good quality comparative studies, many
recommendations are based on lower levels of evidence such as case series. Where
empirical evidence was unavailable, expert opinion relevant to Australia has been
included and stated accordingly.
Plasma-derived factor concentrates remain available to patients who prefer not to
transfer to recombinant products or for those with other rare bleeding disorders that do
2
Use of recombinant and plasma-derived Factor VIII and IX
not have recombinant alternatives. However, due to their higher safety profile
concerning transmission of blood-borne agents, recombinant factors are preferred over
plasma-derived factors. Since studies that evaluated the safety and effectiveness of
plasma-derived products alone did not satisfy the criteria for inclusion in the systematic
review, plasma-derived products were assessed in terms of their relative safety and
effectiveness compared to recombinant products.
Previous guidelines have included the use of porcine factor VIII and aminocaproic acid.
While the effectiveness and safety of porcine factor VIII was evaluated in the systematic
review, all references to the use of porcine factor concentrates have been removed from
the evidence-based guidelines, as it is unavailable in Australia. Similarly, aminocaproic
acid has become unavailable and was not included in these evidence-based guidelines.
Intended use of guidelines
These guidelines are designed as a general guide to inform clinicians on the safest and
most effective treatment strategies using the best evidence available (highest level and
quality of evidence) in published and unpublished literature. They take into consideration
products that are available in Australia, either registered, or available through the
Therapeutic Goods Administration on a ‘named patient’ basis.
These guidelines do not attempt to cover the cost-effectiveness of different treatments.
Despite its rarity, haemophilia is often considered to be the most expensive disease in the
community due to the high costs involved with regular replacement of coagulation
factors VIII and IX and the life-long nature of the disease (Street & Ekert 1996; World
Health Organization 2002). Current treatment may cost up to $110,000 annually per
patient, with clotting factor concentrates comprising 90% of these costs (Waymouth
Resources Limited 2003; World Health Organization 2002). The Australian Health
Ministers’ Advisory Council, Blood and Blood Products Committee Working Party
(AHMAC 2003) recently estimated that the cost of meeting the recommended quantity
and mix of coagulation products in Australia would be $91,950,000 per annum. This
estimate was calculated prior to the recent government decision to fund unrestricted
access to recombinant products for people with haemophilia (Department of Health and
Ageing 2004). Therefore, since recombinant products are approximately 20–50% more
expensive than plasma-derived concentrates (Ananyeva et al. 2004), the annual costs are
likely to increase.
Due to the high cost of products required for the treatment of coagulation disorders, it is
important that resources be used optimally in the safest and most effective manner.
Individual treatment strategies should recognise both patient choice and the availability
of resources at the time.
General Comments
For each individual case of haemophilia, von Willebrand disease or other rare
coagulation disorders, the management of the disease should be associated with a
haemophilia treatment centre (see Appendix C, page 160). All aspects of haemophilia
disease management, including rheumatology, orthopaedic surgery, dentistry, clinical
genetics, infectious diseases, physiotherapy and gynaecology should be managed in
consultation with a haemophilia specialist.
Use of recombinant and plasma-derived Factor VIII and IX
3
Different treatment options should be offered to the patient including information about
the advantages and risks of each choice, so that informed decisions may be made.
To minimise risks associated with treatment products, patients should be vaccinated
against hepatitis A and B if they have not been previously. Close monitoring of any
adverse events such as viral infections or inhibitor formation is essential, particularly after
a change in management.
Due to the lack of higher level evidence in some sections of the systematic literature
review, ongoing randomised controlled trials should be closely monitored for results and
changes to recommendations should be made when new, good quality evidence becomes
available.
4
Use of recombinant and plasma-derived Factor VIII and IX
Haemophilia A, without inhibitors
Products available
•
Recombinant factor VIII concentrates (rFVIII)
•
Plasma-derived factor VIII concentrates (pdFVIII)
•
Desmopressin (1-deamino-D-arginine vasopressin, DDAVP)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
Treatment of acute bleeding episodes
•
Recombinant FVIII is safe and effective for patients without inhibitors to FVIII
(Level II evidence for effectiveness, Level III-2 evidence for safety, page 52).
•
Inhibitor testing, particularly in young children and/or those with severe
haemophilia, should be performed no earlier than day three after initial
administration of factor concentrate, or when the expected response is absent, with
re-testing at regular intervals (Level IV evidence, page 57).
•
Patients with mild/moderate haemophilia A may be treated using DDAVP, at a dose
of 0.3 µg/kg diluted in 50ml of 0.9% saline and infused over ≥30 minutes (Level IV
evidence, page 38).
•
DDAVP may be administered intravenously or subcutaneously with comparable
results (Level II evidence, page 38).
•
DDAVP may be administered once every 24 hours. If given for more than three
consecutive days, repeated doses may lead to a reduction in responsiveness
(tachyphylaxis) (Level IV evidence, page 38). If DDAVP is given more than once in
24 hours, predose monitoring of electrolyte concentrations is recommended (expert
opinion).
•
A test dose of DDAVP and a FVIII/von Willebrand factor (FVIII/vWF) assay
should be performed to demonstrate efficacy (Level IV evidence, page 38).
•
Caution should be taken to restrict fluid intake during DDAVP treatment to prevent
fluid overload (Level IV evidence, page 42). DDAVP should be used with caution in
the elderly; and it is not recommended in those with arteriovascular disease and in
young children (<2 years) (expert opinion).
Use of recombinant and plasma-derived Factor VIII and IX
5
Prophylaxis
•
Recombinant FVIII is recommended for prophylaxis in patients without inhibitors
to FVIII (Level IV evidence, page 78).
•
Prophylaxis should be initiated after bleeding episodes have commenced, rather than
at diagnosis, due to potential increased risk of inhibitor development associated with
administration of factor concentrates in very young children (Level IV evidence,
page 82).
•
Central venous access devices may assist in the regular infusion of factor
concentrates in children. However, adequate aseptic technique should be taught and
routinely reviewed to avoid infection (Level IV evidence). External devices should be
avoided due to the higher rate of infection compared to fully implanted devices
(Level IV evidence, page 132).
Surgical and dental procedures
•
Recombinant FVIII is safe and effective for haemophilia A patients, before or after
surgery as required (Level III-2 evidence for safety and Level IV evidence for
effectiveness, page 106).
•
Tranexamic acid can be used as secondary prophylaxis for surgical or dental
procedures (Level IV evidence, page 108).
•
Consensus-based recommendations for the management of patients undergoing
surgery (Association of Hemophilia Clinic Directors of Canada 1995a) or dental
procedures (Stubbs & Lloyd 2001) have been outlined in detail (Table 37 and
Appendix G). The available evidence that was evaluated in this review is consistent
with the recommendations in these existing guidelines.
6
Use of recombinant and plasma-derived Factor VIII and IX
Haemophilia A, with inhibitors
Products available
•
Recombinant factor VIII (rFVIII)
•
Plasma-derived factor VIII (pdFVIII)
•
Recombinant factor VIIa (rFVIIa) (e.g. NovoSeven)
•
Activated prothrombin complex concentrates (aPCCs) (e.g. FEIBAa)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
Treatment of acute bleeding episodes
•
Recombinant FVIIa is recommended for acute bleeding episodes in patients with
high titre and/or high responder inhibitors to FVIII (Level II evidence for
effectiveness, Level IV evidence for safety, page 90).
•
Recombinant FVIIa may be infused as a bolus at a dose of 90 µg/kg for adults
(Level IV evidence). An appropriate dose of rFVIIa may be given by continuous
infusion as an alternative method of delivery (Level IV evidence, page 90). Paediatric
doses of up to 200–250 µg/kg may be required due to the shorter half-life in children
(expert opinion).
•
There were no available studies with data on the effectiveness of high dose FVIII
concentrates alone in patients with inhibitors (page 90). Therefore, recommendations
for its use are based on consensus only. Expert opinion indicates that there are no
adverse effects at high doses of FVIII.
•
Activated prothrombin complex concentrates (aPCCs) may be used to control mildsevere bleeding in patients with high titre inhibitors. However, patients should be
closely monitored for adverse reactions that, though rare, may be serious. Standard
dose of FEIBAa is 60–100 IU/kg twice per day to a maximum daily dose not
exceeding 200 IU/kg (Level II evidence, page 90).
•
Plasmapheresis (with or without immunoadsorption) may be used to reduce
inhibitors in high titre/high responders before infusion with FVIII to control
bleeding. However, patients should be monitored for potential anaphylactic reactions
(Level IV evidence, page 90).
a
FEIBA = Factor eight inhibitor bypassing agent
Use of recombinant and plasma-derived Factor VIII and IX
7
•
Immunosuppression therapy, using cyclophosphamide, to reduce inhibitors is not
recommended for treating acute bleeding episodes (Level IV evidence, page 90).
Surgical and dental procedures
•
Recombinant FVIIa may be considered the first line of treatment for dental and
other surgical procedures in patients with high titre and/or high responder inhibitors
(Level II evidence for effectiveness and Level IV evidence for safety, page 92).
•
Evidence for the use of recombinant FVIII concentrates for surgery or dental
procedures is currently awaited.
•
Plasma-derived FVIII concentrate may be considered for use during surgery in
patients with low titre, low-responding inhibitors, but should be avoided in patients
with high-responding inhibitors (Level IV evidence, page 92).
•
Activated PCCs may be used for surgical and dental procedures. However, patients
should be closely monitored for thrombotic complications, including disseminated
intravascular coagulation and thrombophlebitis (Level IV evidence, page 92).
•
aPCCs should not be used in conjunction with antifibrinolytic agents, but they may
be used in succession (Level IV evidence, page 92).
Tolerisation procedures
In Australia, tolerisation protocols vary widely across haemophilia centres. There is little
published evidence of effectiveness, no consensus on procedures and the Bonn and
Malmö protocols are not used routinely in Australia. The International Immune
Tolerance Induction study, which includes several participating Australian haemophilia
centres, is ongoing and results from this study, when available (expected completion in
2007), should provide guidance to tolerisation procedures.
Bonn protocol (modified): Patients are given 150 IU/kg FVIII every 12 hours until the inhibitor level
drops below 1 B. Then FVIII concentrate is reduced to 150 IU/kg daily, until inhibitor is no longer
detectable.
•
8
The Bonn protocol may be considered for young patients (Level IV evidence, page
95). Treatment is more likely to be successful with low pre-treatment inhibitor titres,
and with less time between development and treatment of inhibitors (Level IV
evidence, page 95).
Use of recombinant and plasma-derived Factor VIII and IX
Malmö protocol: When inhibitor titres ≥10 BU, extracorporeal immunoadsorption is given until levels
are reduced to <10 BU. High doses of FVIII (approximately 200 IU/kg/day) are administered to
bring the FVIII concentration to 40-100 IU/dL and to maintain it at 30-80 IU/dL. At the same
time, cyclophosphamide is administered at a dose of 12-15 mg/kg intravenously for 2 days, followed by
2-3 mg/kg for 8 days. On day 1 of treatment, immunoglobulin is also used at a dose of 2.5-5 g/kg,
followed by 0.4g/kg on days 4-8. Factor VIII is administered until the inhibitor disappears. Patients
then receive FVIII for ongoing prophylaxis.
•
The Malmö protocol may be considered when patients have long-standing inhibitors
(Level IV evidence, page 95).
Use of recombinant and plasma-derived Factor VIII and IX
9
Haemophilia B, without inhibitors
Products available
•
Recombinant factor IX (rFIX) (BeneFIX®)
•
Plasma-derived factor IX (pdFIX) (MonoFIX®)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
Treatment of acute bleeding episodes
•
Recombinant FIX (BeneFIX®) is safe and effective for the treatment of bleeding
episodes in haemophilia B patients without inhibitors (Level IV evidence, page 67).
Since there is a risk of anaphylaxis occurring within the first 50 days of treatment
with rFIX, patients should be closely monitored and early exposure to factor
concentrate should occur in a centre equipped to treat anaphylaxis (expert opinion).
•
Due to large inter-patient variability, individual dosing regimens should be monitored
by FIX recovery assays (Level II evidence, page 65).
•
The dosage ratio for rFIX to pdFIX is 1.6:1 for patients aged ≤15 years and 1.2:1 for
patients aged 16 years and over (Level III-3 evidence, page 65).
Prophylaxis
•
Recombinant FIX is safe and effective for prophylaxis in haemophilia B patients
without inhibitors (Level II evidence, page 78).
•
Dosing regimens should take into account the lower rFIX recovery compared to
pdFIX and the inter-patient variability—see previous recommendations for
treatment of bleeding episodes (Level II evidence, page 65).
•
Central venous access devices may assist in the regular infusion of factor
concentrates in children. However, adequate aseptic techniques should be taught and
routinely reviewed to avoid infection (Level IV evidence, page 132). External devices
should be avoided due to the higher rate of infection compared to fully implanted
devices (Level IV evidence, page 132).
10
Use of recombinant and plasma-derived Factor VIII and IX
Surgical and dental procedures
•
Recombinant FIX is recommended for haemophilia B patients, before or after
surgery as required (Level IV evidence for safety and effectiveness, page 106).
•
Tranexamic acid can be used as secondary prophylaxis for surgical or dental
procedures (Level IV evidence, page 108).
Use of recombinant and plasma-derived Factor VIII and IX
11
Haemophilia B, with inhibitors
Products available
•
Recombinant factor VIIa (rFVIIa) (e.g. NovoSeven)
•
Activated prothrombin complex concentrates (aPCCs)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
•
Recombinant factor IX (rFIX) (BeneFIX®)
Treatment of acute bleeding episodes
•
Recombinant FVIIa is safe and effective for acute bleeding episodes in patients with
high titre and/or high responder inhibitors to FIX (Level II evidence, page 90).
•
Activated prothrombin complex concentrates (aPCCs) may be considered to control
mild-severe bleeding in patients with high titre inhibitors. However, patients should
be closely monitored for adverse reactions that, though rare, may be serious (Level
IV evidence, page 90).
Tolerisation procedures
•
12
There is no evidence to guide tolerisation procedures in patients with haemophilia B
with inhibitors. Recombinant FIX may be used for tolerisation protocols, due to the
higher safety profile pertaining to transmission of blood-borne agents, but there
should be close monitoring to prevent anaphylactic reactions (expert opinion).
Use of recombinant and plasma-derived Factor VIII and IX
von Willebrand disease
Products available
•
Desmopressin (1-deamino-D-arginine vasopressin, DDAVP)
•
Plasma-derived factor VIII/von Willebrand factor concentrates (pdFVIII/vWF)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
Treatment of acute bleeding episodes
•
Patients with mild von Willebrand disease (vWD) may be treated using DDAVP at a
dose of 0.3 µg/kg diluted in 50ml of 0.9% saline and infused over ≥30 minutes
(Level IV evidence, page 38).
•
DDAVP may be administered intravenously or subcutaneously with comparable
results (Level II evidence, page 38).
•
DDAVP may be administered once every 24 hours. If given for more than three
consecutive days, repeated doses may lead to a reduction in responsiveness
(tachyphylaxis) (Level IV evidence, page 38). If DDAVP is given more than once in
24 hours, predose monitoring of electrolyte concentrations is recommended (expert
opinion).
•
DDAVP should be used only as an adjunct to factor replacement therapy in patients
with severe vWD and not as the primary treatment (Level II evidence, page 38).
•
A test dose of DDAVP and a FVIII/von Willebrand factor (FVIII/vWF) assay
should be performed to demonstrate efficacy (Level IV evidence, page 38).
•
Caution should be taken to restrict fluid intake during DDAVP treatment to prevent
fluid overload (Level IV evidence, page 42). DDAVP should be used with caution in
the elderly; and it is not recommended in those with arteriovascular disease and in
young children (<2 years) (expert opinion).
•
Patients unresponsive to (or contraindicated for) DDAVP should receive
pdFVIII/vWF concentrate, which may be administered every 8–12 hours (Level IV
evidence, page 72).
Use of recombinant and plasma-derived Factor VIII and IX
13
Prophylaxis
•
Prophylaxis may be required for patients with menorrhagia. Limited published
evidence suggests that intranasal DDAVP is no better than placebo at reducing
subjectively measured menorrhagia (Level II evidence, page 85). Plasma-derived
FVIII/vWF concentrates or tranexamic acid are possible alternatives (Level IV
evidence, page 71).
Surgical and dental procedures
•
DDAVP, which may be used in conjunction with local therapies such as fibrin glue
and tranexamic acid, may be considered for use prior to dental extractions or surgery
in patients who respond to DDAVP (test dose) (Level IV evidence, page 111).
•
Plasma-derived FVIII/vWF concentrates may be used to cover surgical or dental
procedures (Level IV evidence, page 112).
14
Use of recombinant and plasma-derived Factor VIII and IX
Other rare bleeding disorders
Products available
•
Recombinant factor VIIa (rFVIIa)—Factor VII deficiency
•
Prothrombin complex concentrates—Factor II, IX, or X deficiency
•
Fibrinogen—Fibrinogen deficiency
•
Plasma-derived factor VII—Factor VII deficiency
•
Plasma-derived factor XI—Factor XI deficiency
•
Plasma-derived factor XIII—Factor XIII deficiency (e.g. Fibrogammin P)
•
Cryoprecipitate—Factor XIII deficiency, afibrinogenaemia, dysfibrinogenaemia
•
Fresh frozen plasma—Factor V deficiency
•
Adjunctive agents (e.g. fibrin glue)
Use of recombinant and plasma-derived Factor VIII and IX
15
Acquired haemophilia
Products available
•
Desmopressin (1-deamino-D-arginine vasopressin, DDAVP)
•
Activated prothrombin complex concentrates (aPCCs)
•
Recombinant factor VIIa (rFVIIa) (e.g. NovoSeven)
•
Immunosuppressants (e.g. rituximab)
•
Antifibrinolytics (e.g. tranexamic acid)
•
Adjunctive agents (e.g. fibrin glue)
Treatment of acute bleeding episodes
•
Bleeding episodes in patients with acquired haemophilia require prompt treatment
(Level IV evidence, page 99).
Inhibitors <5 BU
•
DDAVP should be used at the dose recommended above (Haemophilia A, without
inhibitors). Plasma-derived factor VIII concentrate may be used at a dose of 20
IU/kg for each BU of inhibitor plus an additional 40 IU/kg and tested after 15
minutes. If response is poor, another bolus injection may be given (Level IV
evidence, page100).
Inhibitors >5 BU
•
Activated PCCs may be administered at a dose of 50–200 IU/kg/day in divided
doses (Level IV evidence, page 100).
•
Recombinant FVIIa may be administered at a dose of 90 µg/kg every 2–6 hours
until the bleeding stops (Level IV evidence, page 100).
•
Due to the risk of thrombotic events, patients should be closely monitored after
treatment (Level IV evidence, page 99).
Tolerisation protocols
•
Patients with acquired haemophilia should be treated for the underlying disorder
before deciding on strategies to eliminate inhibitors (Level IV evidence, page 99).
Postpartum: Patients may be monitored, without treatment, as inhibitors most
frequently disappear spontaneously. Alternatively, immunosuppressive therapy may
be considered (Level IV evidence, page 98).
16
Use of recombinant and plasma-derived Factor VIII and IX
Drug-related: The drug should be withdrawn to allow spontaneous disappearance of
inhibitors. Alternatively, immunosuppressive therapy may be considered (Level IV
evidence, page 99).
Autoimmune disease: Immunosuppressive therapy should be given, as inhibitors are
unlikely to disappear spontaneously (Level IV evidence, page 98).
Malignant neoplasm: Patients should be treated for the primary malignancy.
Immunosuppressive therapy should then be given (Level IV evidence, page 99).
Idiopathic: Patients should be given immunosuppressive therapy (Level IV evidence,
page 100).
Use of recombinant and plasma-derived Factor VIII and IX
17
Infant delivery
Unless stated, all recommendations are based on Level IV evidence. Recommendations
assume a correct diagnosis and that the severity of the disorder has been ascertained.
Antenatal care
•
Counselling pertaining to the potential risks of antenatal testing and the potential
complications during pregnancy and delivery should be accessible to all haemophilia
carriers and women with von Willebrand disease (page 118).
•
If antenatal testing is accepted, chorionic villus sampling is the preferred method. If
the levels of FVIII or FIX are low prior to pregnancy, FIX levels in haemophilia B
carriers and FVIII levels in haemophilia A carriers should be monitored, particularly
in the third trimester and postpartum (page 120).
Delivery of infants
•
Invasive fetal monitoring, such as scalp electrodes, instrument deliveries and long
labours should be avoided for affected infants (page 121).
•
Vaginal delivery is the recommended mode of delivery, unless obstetric factors
indicate caesarean section. Vacuum extraction delivery is contraindicated (page 121).
•
Use of DDAVP in women with von Willebrand disease is not recommended during
labour (expert opinion, page 38).
Postnatal and postpartum care
•
Cord or peripheral blood samples should be taken from affected infants of
haemophilia carriers and infants with suspected bleeding disorders to ascertain
coagulation factor status (page 121).
•
All infants with intracranial haemorrhage should be evaluated for the presence of a
bleeding disorder, even where there is no family history of haemophilia. DDAVP is
contraindicated in neonates (page 121).
•
Heel sticks and venipuncture, other than to assess coagulation factor status, should
be avoided if possible. Intramuscular vitamin K is associated with bleeding in infants
with haemophilia. Oral vitamin K is available as an alternative (page 121).
•
Fibrin glue may be used in conjunction with pdFVIII/vWF or pdFIX concentrates
to achieve haemostasis if circumcision is performed (Level IV evidence, page 50).
The use of rFVIII or rFIX has not been assessed in this context.
•
DDAVP may be used for prophylaxis during the first 3–4 days after delivery in
women with mild Type 1 von Willebrand disease to increase and maintain factor
levels (page 38).
18
Use of recombinant and plasma-derived Factor VIII and IX
•
All Type 2 and 3 von Willebrand patients who fail to reach optimal FVIII:C levels
may be treated with pdFVIII/vWF concentrates postpartum (page 125).
Use of recombinant and plasma-derived Factor VIII and IX
19
Background
Normal haemostasis
When blood vessels are damaged through injury, there are three key mechanisms that
promote haemostasis—i.e. to stop any bleeding that occurs. Vasoconstriction, which
occurs within seconds following loss of vascular integrity, is the initial mechanism that
reduces the flow of blood to the site of injury. This is followed by platelets adhering to
the site of the injured vessel wall, forming an aggregate or haemostatic plug. Adhesion of
the platelets is mediated by von Willebrand factor, a protein that binds to and stabilises
blood coagulation factor VIII (Braunwald et al. 2001). The third phase, which takes place
over several minutes, involves a cascade of reactions that culminate in the production of
sufficient thrombin to convert fibrinogen to fibrin—a key element in strengthening the
haemostatic plug and forming a fibrin clot. Figure 1 illustrates a schematic model of the
coagulation cascade. Over 20 proteins are involved in the complete coagulation process
and two of these, factors VIII and IX, are central to this process. These two factors
activate FX to FXa, a necessary component in the activation of prothrombin to
thrombin. Thrombin, in turn, converts fibrinogen to fibrin, which attaches to the platelet
plug, forming a fibrin mesh, or blood clot.
Injury
IX
IX
Tissue
factor
pathway
inhibitor
VIIa
Tissue factor
XI
Thrombin
activation
X
Xa
Va
Prothrombin
Figure 1.
Thrombin
Fibrinogen
(a=activated)
XIa
IXa
VIIIa
Fibrin
A schematic model of blood coagulation after injury (modified from Bolton-Maggs et al. 2003)
Haemophilia
Haemophilia is an inherited blood coagulation disorder that results from a functional
deficiency in specific blood clotting factors. The most common deficiencies are of factor
VIII (haemophilia A or Classic haemophilia) or factor IX (haemophilia B or Christmas
disease). A deficiency in either factor leads to insufficient activation of thrombin, slower
blood clotting and weak, unstable blood clots (Bolton-Maggs & Pasi 2003). The severity
of haemophilia is inversely related to the level of circulating functional blood clotting
proteins (Table 1).
Use of recombinant and plasma-derived Factor VIII and IX
21
Table 1.
Classification of haemophilia
Concentration of factor (VIII or IX)
Classification
Clinical characteristics
<0.01 IU/ml (<1% of normala)
Severe
Spontaneous joint and muscle bleeding; bleeding after injuries,
accidents, and surgery
0.01 - 0.05 IU/ml (1-5% of normal)
Moderate
Bleeding into joints and muscles after minor injuries; excessive
bleeding after surgery and dental extractions
>0.05 – 0.40 IU/ml (5-40% of normal)b
Mild
Spontaneous bleeding does not occur: bleeding after surgery,
dental extractions, and accidents
Modified from Bolton-Maggs & Pasi 2003; a Normal concentration of factor VIII or IX is defined as 100% or one unit of factor VIII activity per ml
of plasma - 100 U/dL (Kasper 2004); b levels of FVIII above 40% are considered sufficient for normal haemostasis.
The hallmark of haemophilia is bleeding into organs or muscle tissue (haematoma) or
joint spaces (haemarthrosis), which may occur without trauma. Frequent bleeding into
joints thickens synovial tissue and produces more friable blood vessels. This cycle of
repeated bleeding may lead to chronic arthritis and a steady decrease in joint mobility and
function (Bolton-Maggs & Pasi 2003). Haemophilia is also characterised by spontaneous
bruising and bleeding gums (World Health Organization 2002). Given that haemophilia
A and B are clinically indistinguishable, a diagnosis must be confirmed by a specific
factor assay (Hitchens 2004).
Since both haemophilia A and B are determined by recessive mutant genes on the Xchromosome, they predominantly affect males (Hitchens 2004). A female may be a
carrier, but have no symptoms of haemophilia as she has another X-chromosome to
produce factor VIII and IX. However, a proportion of female carriers, who have very
low concentrations of factors, may be predisposed to excessive bleeding, demonstrating a
form of haemophilia (Bolton-Maggs & Pasi 2003). In rare cases, if the relevant genes on
the normal X-chromosome are switched off (lyonisation), females may develop a more
severe form of haemophilia (Kulkarni & Lusher 2001).
Haemophilia is diagnosed most commonly in those with a known family history of
haemophilia, or following presentation with bleeding. Although most infants with severe
haemophilia are born without incident, intracranial haemorrhage during the neonatal
period occurs in 1–4% of cases, particularly after vacuum extraction (Bolton-Maggs &
Pasi 2003).
Acquired haemophilia, which is caused by the development of inhibitors to factor VIII in
people without a family or personal history of bleeding diathesis, is characterised by the
sudden onset of bleeding, usually severe (Baudo et al. 2004; Delgado et al. 2003). In 40–
50% of cases the aetiology is unknown, whereas the remainder are associated with
conditions such as pregnancy, autoimmune disease, malignant neoplasm and drug
administration (Delgado et al. 2003).
Burden of disease
Haemophilia A accounts for approximately 80% of haemophilia while the remaining
20% of people with haemophilia have haemophilia B (Haemophilia Foundation Australia
2004b). Currently, haemophilia affects approximately 1800 people in Australia (one in
7000 males) (Haemophilia Foundation Australia 2004b). In 2001–02, there were 912
22
Use of recombinant and plasma-derived Factor VIII and IX
hospital separationsb for haemophilia A and 59 hospital separations for other hereditary
coagulation factor deficiencies in Australia (AIHW 2004). Approximately one third of
cases are thought to have no known family history of haemophilia (Chalmers 2004). In
addition, acquired haemophilia affects approximately one in a million people (Baudo et
al. 2004; Delgado et al. 2003).
The prevalence of haemophilia in the community is likely to rise with the increasing
longevity of people with haemophilia. For example, in 2000, data from the National
Bleeding Disorder Registry (which includes all Australian states except NSW) showed
that nearly 45% of people with severe haemophilia A and 50% of those with severe
haemophilia B were aged 18 or under (AHMAC 2003). Compared to previous cohorts
who had a higher risk of transmission of infectious diseases via blood products that were
not adequately screened or heat treated, the life expectancy for current (and future)
cohorts of people with haemophilia is greater, leading to a potential increase in the
demand for clotting factors.
Treatment regimens
Since there is no cure for haemophilia at the present time, standard treatment for
bleeding episodes involves replacing the deficient factors (VIII or IX). Before treatment
with appropriate blood clotting factors was available, people with haemophilia had a
short life expectancy and were likely to die prematurely from a spontaneous bleeding
episode (World Health Organization 2002). A range of different factor replacement
methods involving intravenous administration of fresh or commercial factor
concentrates has been developed. People with severe haemophilia are likely to receive
approximately 14,000 intravenous infusions of factors in a lifetime. Administration of
specific doses, adjunctive haemostatic agents, and additional medical intervention
depends on the site and severity of the bleeding episode.
The two main forms of treatment for haemophilia are ‘on-demand’ or ‘prophylactic’, and
factor concentrates may be either plasma-derived or recombinant (genetically engineered
product that does not require human blood). For on-demand treatment, patients are
treated only when a bleeding episode occurs, either spontaneously or associated with
trauma (World Health Organization 2002). Prophylactic treatment is based on the
principle of maintaining clotting factor levels above 1% of normal, to convert the
bleeding pattern of severe haemophilia to that found in milder levels of the disease and
involves regular factor infusions regardless of bleeding status (Nilsson & Hedner 1976).
Prophylactic infusions are also administered prior to surgical or dental procedures.
von Willebrand disease
Von Willebrand disease is caused by a deficiency (either quantitative or functional) in von
Willebrand factor (vWF). This factor circulates in the blood, acting as a carrier protein to
factor VIII and as an adhesive protein involved in platelet-vessel wall interaction (Laffan
et al. 2004). Deficiency of vWF causes inadequate platelet adhesion and secondary
deficiency of factor VIII. It is characterised by easy bruising, bleeding from mucous
b
Hospital separations = a hospital transfer, discharge or death (Australian Bureau of Statistics)
Use of recombinant and plasma-derived Factor VIII and IX
23
membranes, post-operative bleeding and menorrhagia (heavy menstrual bleeding). Unlike
haemophilia A and B, which are determined by mutations on the sex chromosome, von
Willebrand disease affects males and females equally (Laffan et al. 2004). There are three
main types of von Willebrand disease, depending on whether the vWF is partially
deficient (Type 1), dysfunctional (Type 2) or completely absent (Type 3, a rare autosomal
recessive form) (Laffan et al. 2004). The most common symptoms of von Willebrand
disease are epistaxis (bleeding from the nose) and postoperative bleeding in men and
women and menorrhagia in women (Ziv & Ragni 2004).
Burden of disease
Approximately 1016 people in Australia have been diagnosed with von Willebrand
disease (Australian Bleeding Disorder Registry 2004) (personal communication, Simone
Proft, Population Health Division, Department of Health and Ageing). However, since it
generally presents in its mild or moderate form, von Willebrand disease frequently goes
undiagnosed and untreated (Haemophilia Foundation Australia 2004b). In 2001–02 there
were 342 hospital separations for people with von Willebrand disease in Australia
(AIHW 2004).
Treatment regimens
Mild to moderate forms of von Willebrand disease (Type 1) are treated primarily with
desmopressin (DDAVP), an analogue of vasopressin that stimulates release of von
Willebrand factor (vWF) into the plasma. Bleeding episodes in severe Type 1 and in Type
2 and 3 von Willebrand disease are treated by infusion of plasma-derived factor VIII
concentrate containing vWF (pdFVIII/vWF concentrate) (Braunwald et al. 2001).
Other rare bleeding disorders
While haemophilia A, B and von Willebrand disease represent the larger proportion of
inherited bleeding disorders, a series of rare blood coagulation disorders, which are
associated with deficiencies in other clotting factors, make up the balance.
A deficiency in factor V (otherwise known as Owren’s disease or parahaemophilia, and
distinct from factor V Leiden) in its homozygous state affects approximately one in a
million people and affects men and women equally. It is usually associated with easy
bruising and mucous membrane bleeding, such as epistaxis or oral cavity bleeding
(Bolton-Maggs et al. 2004).
Clotting factor X, or Stuart-Prower factor, is an enzyme in the coagulation pathway and
deficiency in this factor may be inherited (autosomal recessive) or acquired (associated
with severe liver disease, vitamin K deficiency, or anticoagulant drugs such as warfarin)
(Steen & Schwartz 2004). Symptoms of factor X deficiency include severe umbilical
stump bleeding in newborn infants, epistaxis, menorrhagia, easy bruising, recurrent
miscarriage, postpartum bleeding and excessive bleeding following surgery or trauma.
Inherited factor VII deficiency is a rare autosomal recessive bleeding disorder, which
results from mutations in the FVII gene located on chromosome 13 and affects males
and females equally (Israels 2004a). Factor VII is a vitamin K-dependent coagulation
24
Use of recombinant and plasma-derived Factor VIII and IX
factor with a short half-life (3–4 hours). Deficiency or dysfunction of this factor is
characterised by mucosal bleeding, such as epistaxis and menorrhagia, post-operative
bleeding and intracranial haemorrhaging in the first year of life.
Deficiency in factor XI (also known as haemophilia C) is a milder bleeding disorder, with
autosomal inheritance, that occurs predominantly in Ashkenazi Jews (Bolton-Maggs &
Pasi 2003). Depending on the severity of the deficiency, the first sign of this bleeding
disorder may occur at circumcision, menarche, or during dental extractions, trauma or
surgery (Siegel 2004).
Inherited factor XIII deficiency is a rare autosomal recessive disease primarily caused by
mutations in the gene located on chromosome 6 (Israels 2004b). Factor XIII is required
for the final step in the coagulation cascade and stabilisation of the fibrin plug. Factor
XIII deficiency is often detected within the first few days of life due to bleeding from the
umbilical cord stump. Common characteristics of this disorder include intracranial
haemorrhage in the infant, bruising and soft tissue bleeding, delayed bleeding after
trauma, recurrent miscarriages and poor wound healing.
Prothrombin deficiency is an extremely rare autosomal recessive disorder, with
approximately 26 cases reported worldwide (Di Paola et al. 2001). Acquired prothrombin
deficiency, which is more common, has been associated with severe liver disease, use of
prescription drugs to inhibit blood clotting (e.g. warfarin) and vitamin K deficiency
(Canadian Hemophilia Society 2004). Prothrombin, or factor II, is converted by factor
Xa to thrombin, an essential component in the clotting cascade (see Figure 1).
Characteristics of prothrombin deficiency include umbilical cord stump bleeding at birth,
epistaxis, easy bruising, menorrhagia, postoperative and/or postpartum bleeding and
occasional muscle haematomas.
Complications of treatment
The primary complications associated with treatment for haemophilia are transmission of
viral infections, such as human immunodeficiency virus (HIV), hepatitis B and C and the
theoretical risk of prion transmission through infected pooled plasma concentrates, and
the development of inhibitors to the clotting factors (Street & Ekert 1996). Between
1981 and 1984, 245 people with haemophilia contracted HIV in Australia and the
majority of patients who received factor concentrates before 1989 were infected with
hepatitis C. After 1989 the safety profile of blood products from plasma improved.
Technologies became available to heat treat the plasma products to inactivate bloodborne viruses and donors were screened for HIV and hepatitis C (from 1990) (Street &
Ekert 1996; Haemophilia Foundation Australia 2004a). Despite recent advances in
creating high purity plasma-derived concentrates there is still anxiety in the haemophilia
community about contracting viruses not yet identified through infusions of plasmaderived factors (Haemophilia Foundation Australia 2004a). For example, there remains a
theoretical risk of transmission of prions, such as Creutzfeldt-Jakob disease. With the
introduction of blood donor screening, donor testing and virus inactivation techniques,
there have been no reported events of viral transmission from infected blood
transfusions since the early 1990s. At present, however, the risk of transmission of prions
is unknown in Australia.
Another serious complication of treatment with clotting factors (both plasma-derived
and recombinant) is the development of inhibitory antibodies. This occurs when the
Use of recombinant and plasma-derived Factor VIII and IX
25
body recognises the therapeutic clotting agents as foreign proteins (Giangrande 1996).
Up to one third of people with severe haemophilia A develop neutralising antibodies, or
inhibitors, while the incidence in those with haemophilia B is 1–6% (World Health
Organization 2002). There are significant clinical implications associated with inhibitor
development for a proportion of patients with inhibitors, as the response to treatment
becomes uncertain, morbidity increases, and life expectancy is reduced (Hay et al. 2000b).
The direct and indirect costs to patients, their families and society increase. To
counteract the effects of inhibitor development, a tolerisation procedure may be
introduced. Tolerisation (or immune tolerance induction) may be achieved by the regular
administration of factor VIII or IX over a period of months or years.
26
Use of recombinant and plasma-derived Factor VIII and IX
Approach to Guideline Development
How do we develop clinical practice guidelines?
Evidence-based clinical practice guidelines are developed as part of a systematic literature
review.
‘Systematic reviews provide information about the effectiveness of interventions by
identifying, appraising, and summarising the results of otherwise unmanageable quantities
of research.’ (Khan et al. 2001) The primary focus of systematic reviews in medicine is to
integrate empirical research for the purpose of creating generalisations and thus to
provide a rational basis for health care decision making and the development of
guidelines for clinical practice (Mulrow et al. 1997; Cooper & Hedges 1994).
The key components of a systematic review include: 1) the development of specific
research questions or hypotheses; 2) a transparent methodical process defined a priori (i.e.
a review protocol); 3) an exhaustive search for relevant primary research on the topic; 4)
the critical appraisal of this research; 5) an attempt to answer the research questions and
to resolve conflicts in the literature; and 6) the identification of issues central to future
research on the topic (Mulrow et al. 1997; Cooper & Hedges 1994; Clarke & Oxman
2000). Evidence that has been synthesised can then inform the development of a
guideline pertaining to each clinical practice question.
In the light of recent policy initiatives by the Australian Government (Department of
Health and Ageing 2004), the main objectives of this guideline document were to
develop: 1) national clinical practice guidelines for the use of recombinant factors VIII
and IX for patients with Haemophilia A and B; 2) national clinical guidelines for the use
of plasma-derived factors for patients with von Willebrand Disease; and 3) national
tolerisation protocols for tasks 1) and 2) above.
Since the Report of the Working Party on the supply and use of Factor VIII and Factor IX in
Australia (AHMAC 2003) recently examined the clinical need and costs of treatment in
Australia, this guideline document omits any systematic determination of clinical need
and cost-effectiveness.
Although the systematic literature review that underpins the development of these
clinical practice guidelines examined the effectiveness and safety of recombinant
compared to plasma-derived factors VIII and IX, the new clinical practice guidelines
have incorporated recent government policy to provide full access to recombinant
factors VIII and IX for haemophilia A and B patients. The evidence-based
recommendations also focus on the implications of transferring patients from plasmaderived to recombinant products. Other guidelines sections, which remain unaffected by
the new policy, include the use of plasma-derived factors for patients with von
Willebrand disease, the use of adjunctive haemostatic agents, and the development of
national tolerisation protocols for patients with haemophilia A or B.
Use of recombinant and plasma-derived Factor VIII and IX
27
Research questions
The main research questions that this report was commissioned to investigate are:
1.
What is best practice for the treatment of acute bleeding episodes in patients with
haemophilia A, B, von Willebrand disease and other rare coagulation disorders?
2.
What is best practice for the prophylactic treatment of patients with haemophilia A,
B or von Willebrand disease?
3.
What is the best method for treating bleeding episodes in patients with inhibitors to
factors VIII or IX?
4.
What is best practice for the implementation of tolerisation procedures for patients
with inhibitors to factors VIII or IX?
5.
What is the best method of managing patients with haemophilia A, B, von
Willebrand disease and other rare coagulation disorders undergoing surgical and
dental procedures?
6.
What is best practice in the delivery of infants with haemophilia A or B and the
management of infant delivery in women with von Willebrand disease and other rare
coagulation disorders?
Overview of methodology
Inclusion criteria
Criteria for including studies in this systematic review and clinical practice guidelines
document are provided in Box 1 (Appendix E). In order to ensure that the selection of
studies was not biased, these criteria were delineated prior to collating the literature.
Search strategy
Prevalence data on haemophilia and von Willebrand disease were obtained from the
Australian Bleeding Disorder Registry (Australian Bleeding Disorder Registry 2004) and
the Population Health Division, Department of Health, NSW.
The medical and health literature was searched to identify relevant studies and reviews to
answer the research questions on the management and treatment of patients with
haemophilia A, B, von Willebrand disease and other rare blood coagulation disorders.
The search period was from 1966 to September 2004. Table 56 (Appendix E) lists the
electronic databases that were used for this search. Table 57 (Appendix E) lists other
potentially relevant sources of literature that were canvassed, including grey literature.
The search terms used for identification of literature on haemophilia A, haemophilia B
and von Willebrand disease, and concerning factors VIII and IX, were developed on an
Embase platform, which includes Embase and Medline databases (Table 58, Appendix
E). Similar strategies were used for other electronic databases, with the same text words
being used along with the relevant alternatives to indexing headings.
28
Use of recombinant and plasma-derived Factor VIII and IX
The full texts of all articles that were deemed potentially relevant (453) were retrieved for
further screening and allocated to different topic areas—haemostatic agents, treatment of
bleeding episodes, prophylaxis, inhibitors and tolerisation protocols, surgical and dental
procedures, infant delivery and other aspects of management. Three researchers judged
the eligibility for inclusion of all retrieved studies according to the inclusion criteria
developed in the review protocol. A description of all the studies that met the inclusion
criteria for assessing the effectiveness and safety of the different therapies is provided in
Appendix I.
Many studies that reported on treatment of patients with haemophilia A, B or von
Willebrand disease failed to meet the inclusion criteria. Exclusion was primarily due to
inappropriate study design (case reports or expert opinion); assessment of efficacy of
plasma-derived products alone in haemophilia A or B; or inability to distinguish between
therapies as patients treated with recombinant and plasma-derived factor were combined
in the same group.
In order to minimise duplication of data extraction, authors, time period and location of
studies, patient characteristics, and methodological details were examined closely. Where
multiple publications reported data on the same patient population, data were extracted
from the paper providing the most complete follow-up. It should be noted, however,
that it was not always clear to what extent data overlapped multiple publications. Time
constraints precluded contacting authors for missing data or clarification of
methodology.
Critical appraisal
The evidence presented in the included studies was assessed and classified using the
dimensions of evidence defined by the National Health and Medical Research Council
(NHMRC 2000a). These dimensions (Table 2) consider important aspects of the
evidence supporting a particular intervention and include three main domains: strength
of the evidence, size of the effect and relevance of the evidence. The three sub-domains
(level, quality and statistical precision) are collectively a measure of the strength of the
evidence. The designations of the levels of evidence are shown in Table 3. Only the
highest level of evidence was reported for each research question.
Table 2.
Evidence dimensions
Type of evidence
Definition
Strength of the evidence
Level
The study design used, as an indicator of the degree to which bias has been eliminated by
design.*
Quality
The methods used by investigators to minimise bias within a study design.
Statistical precision
The p-value or, alternatively, the precision of the estimate of the effect. It reflects the
degree of certainty about the existence of a true effect.
Size of effect
The distance of the study estimate from the ‘null’ value and the inclusion of only clinically
important effects in the confidence interval.
Relevance of evidence
The usefulness of the evidence in clinical practice, particularly the appropriateness of the
outcome measures used.
*See Table 3
Use of recombinant and plasma-derived Factor VIII and IX
29
Table 3.
Designations of levels of evidence*
Level of evidence
Study design
I
Evidence obtained from a systematic review of all relevant randomised controlled trials
II
Evidence obtained from at least one properly designed randomised controlled trial
III-1
Evidence obtained from well designed quasi-randomised controlled trials (alternate allocation or
some other method)
III-2
Evidence obtained from comparative studies (including systematic reviews of such studies) with
concurrent controls and allocation not randomised, cohort studies, case-control studies, or
interrupted time series with a control group
III-3
Evidence obtained from comparative studies with historical control, two or more single arm studies,
or interrupted time series without a parallel control group
IV
Evidence obtained from case series, either post-test or pre-test/post-test
*Modified from (NHMRC 2000a).
Appraisal of the included papers was undertaken using several checklists appropriate to
each particular study design—systematic reviews (Khan et al. 2001); intervention studies
(Downs & Black 1998); or non-comparative studies, such as uncontrolled before-andafter studies and case series (Young et al. 1999) (Appendix H).
Statistical precision was determined using standard statistical principles. Small confidence
intervals and p-values give an indication of the probability that the reported effect is real
(NHMRC 2000b). Clinically important benefits of the effect size associated with
outcomes and the clinical relevance of the evidence were assessed using appropriate
checklists (Appendix H). A greater than 20% difference, compared to a baseline measure
or the control group performance, was used as an arbitrary measure of clinical
importance.
Data Extraction and Analysis
The process of study selection went through six phases and the number of literature
citations retrieved and retained at each phase was documented (Figure 2, Appendix E).
Evidence tables, as described in Appendix I, were used as a guide to summarise the
extraction of data (NHMRC Guidelines Assessment Register Consultants Working Party
2004). Definitions of specific safety and effectiveness outcomes were provided in the
review protocol. All data extraction was checked by another researcher for face validity.
Information on each of the relevant outcomes was extracted, tabulated and summarised
in the body of the report—including numerator and denominator information, means
and standard deviations, and summary measures of effect, where appropriate. All
statistical calculations and testing were undertaken using the statistical computer package,
Stata version 8.2 (Stata Corporation 2004).
Meta-analysis was not feasible due to the paucity of high level evidence, the
heterogeneous nature of the available randomised crossover trials, and the potential for
duplication of data in multiple publications.
The quantity and quality of evidence varied across the different sections of this report.
For each section, the highest level of evidence is presented and, where possible, data
were extracted from comparative studies. For the most part, however, the evidence base
consisted of low level evidence (case series) and a ‘best evidence’ approach was used.
30
Use of recombinant and plasma-derived Factor VIII and IX
Selection of Products
The key advantage to using recombinant products over plasma-derived products is
believed to be their higher safety profile with a lower transmission of blood-borne
pathogens. Therefore, recombinant products should be used preferentially over plasmaderived products for patients with haemophilia A or B. Since the Australian Government
announced unrestricted access to recombinant factors VIII and IX for all individuals
with haemophilia A or B, this review focuses on the effectiveness, safety and appropriate
use of recombinant factor concentrates for the management of haemophilia A and B. The
effectiveness and safety of other products, such as plasma-derived factor concentrates
and adjunctive haemostatic agents, are also evaluated in relation to their use in the
treatment of patients with von Willebrand disease and other rare coagulation disorders,
or in patients with inhibitors to factors VIII or IX.
Table 4 to Table 9 list products that are registered in Australia and include those that are
available, not currently available, and those available on the Special Access Schemec, for
use in patients with haemophilia A, B, von Willebrand disease and other rare bleeding
disorders. All statements on the therapeutic use of these products are based on Level IV
evidence (Haemophilia Foundation Australia Medical Advisory Panel 2000).
Fresh plasma products
The first successful treatment of haemophilia A, using whole blood, was reported in
1840. In 1929, a technique was developed that enabled plasma to be separated from
whole blood. Until the 1960s whole blood and blood plasma were the only therapeutic
options for people with haemophilia A and B. In 1965, cryoprecipitate was developed for
the treatment of haemophilia A, enabling local blood banks to stock this effective and
portable treatment (Kingdon & Lundblad 2002). Both fresh frozen plasma and
cryoprecipitate are still available for treatment of some blood coagulation disorders (see
Table 4), but safer products have superseded their use for haemophilia A and B.
Table 4.
Fresh plasma products not virally inactivated*
Product
Manufacture of product
Therapeutic use of product
Fresh frozen plasma
Plasma is separated from whole blood
through centrifugation, and then snap frozen
in a mechanical freezer. Fresh frozen
plasma contains approximately 200 units of
FVIII coagulant activity in 150–300ml
volume.
For use in treatment of patients with FV deficiencies.
May be used for FXI deficiency when FXI concentrate
is contraindicated.
Cryoprecipitate
Fresh frozen plasma is thawed over 24
hours at 4°C and the cold insoluble
precipitate is separated through
centrifugation. Cryoprecipitate contains 100
units of FVIII coagulant activity per bag.
For use in some circumstances for vWD and
hypofibrinoginemia patients.
* modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000; FV= factor V; FXI= factor XI; vWD= von Willebrand
Disease
c The Special Access Scheme refers to arrangements that provide for the import and/or supply of an
unapproved therapeutic good for a single patient, on a case-by-case basis.
Use of recombinant and plasma-derived Factor VIII and IX
31
Plasma-derived coagulation factor concentrates
In 1968, the first clotting factor concentrate (Hemofil®) was produced on an industrial
scale. Between 1985 and 1992, all available plasma-derived factor concentrates became
virally inactivated against lipid-enveloped viruses, including hepatitis C, hepatitis B and
HIV (Kingdon & Lundblad 2002). Von Willebrand factor, which is not present in
recombinant or immuno-affinity purified FVIII concentrates, is preserved in the plasmaderived FVIII concentrate, Biostate®. Human plasma-derived factors available in
Australia are listed in Table 5.
Table 5.
Plasma-derived coagulation factor concentrates*
Type of
product
Product
(Manufacturer)
Manufacture of product
Therapeutic use of
product
pdFVIII
Biostate®
(CSL)
Dried factor VIII preparation, which is fractionated from
cryoprecipitate, contains von Willebrand factor. Fibrinogen
and fibronectin are removed by precipitation with heparin and
by gel filtration. Factor VIII is precipitated with glycine sodium
chloride and lyophilised after albumin is added. Viral
inactivation is by solvent detergent plus dry superheating for
72 hours at 80°C. Each vial of FVIII concentrate contains 250
units of FVIII and 500 units of von Willebrand factor: ristocetin
cofactor activity.
For the control and
prevention of haemorrhagic
episodes and for surgical
prophylaxis in patients with
haemophilia A and von
Willebrand Disease.
pdFIX
MonoFIX®-VF
(CSL)
Factor IX concentration is chromatographically purified, with
two viral inactivation or removal steps, solvent detergent and
nanofiltration. Heparin and human antithrombin III are added.
Each vial of FIX concentrate contains 500 units of FIX.
For the treatment of
haemorrhages and for use
in minor surgery and as
prophylaxis in patients with
haemophilia B.
*modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000; pdFVIII= plasma-derived factor VIII; pdFIX= plasmaderived factor IX,
Recombinant coagulation factor concentrates
In 1987, the first recombinant factor VIII product was infused into a haemophilia A
patient. From December 1992, Recombinate™ was available in the United States, and
Kogenate® followed in 1993. In 1999, a B-domain-deleted recombinant factor VIII
concentrate became available in Europe (ReFacto®) and was licensed in the United
States in 2000 (Kingdon & Lundblad 2002). The smaller, more stable B-domain-deleted
recombinant FVIII molecule precludes the need for human albumin as a stabiliser
(Fijnvandraat et al. 1997). Prompted by the desire to improve their efficacy and safety
profile, recombinant factors have developed through several generations, listed in Table
6.
First generation products were manufactured using animal and human proteins in the cell
culture medium and with albumin to stabilise the product. Second generation products
are manufactured without albumin, which has been replaced with a non-protein
stabiliser, such as sucrose. Third generation products do not have human or animal
proteins added to the cell culture. Excluding human or animal products from the
recombinant products reduces the risk of transmission of infectious agents (United
Kingdom Haemophilia Centre Doctors' Organisation 2003).
In 1997, the first recombinant factor IX for haemophilia B was developed (BeneFix®),
based on the technologies used for recombinant factor VIII (Kingdon & Lundblad
2002). Activated recombinant factor VII (rFVIIa, NovoSeven®) is the most recent
development in treatments for haemophilia patients with inhibitors (Kingdon &
Lundblad 2002).
32
Use of recombinant and plasma-derived Factor VIII and IX
Table 6.
Recombinant coagulation factor concentratesa
Type of
product
Product
(Manufacturer)b
Manufacture of product
1st
generation
Recombinate™
(Baxter)
Factor VIII and vWF genes are inserted into Chinese hamster
ovary cell lines. The vWF is used as a stabiliser for FVIII. The
secreted rFVIII is purified by single immunoaffinity
chromatography using a murine monoclonal antibody, and by
two ion-exchange chromatography steps. Purified rFVIII is
stabilised with pasteurised human albumin.
Kogenate® (Bayer),
Helixate® (Bayer)
Factor VIII genes are inserted into baby hamster kidney cell
lines. The secreted rFVIII is purified with two ion-exchange
gel filtration/size exclusion chromatography and double
immunoaffinity chromatography using a murine monoclonal
antibody. Purified rFVIII is stabilised with pasteurised human
albumin and contains trace hamster protein, trace murine
immunoglobulin and trace von Willebrand factor. Viral
inactivation is achieved through heat treatment.
rFVIII
2nd
generation
rFVIII
Therapeutic use of
product
The treatment of
choice for the control
and prevention of
haemorrhagic
episodes and for
routine and surgical
prophylaxis in
patients with
haemophilia A.
ReFacto® (Wyeth
Pharmaceuticals)
B-domain deleted FVIII genes are inserted into Chinese
hamster ovary cell lines. Purification involves five
chromatographic steps and one virus inactivation step by
solvent detergent. Human albumin is not added. Trace
amounts of hamster protein and murine protein are included.
Kogenate® FS
(Bayer), Helixate
Nexgen® (Bayer)
Different to Kogenate, Kogenate FS- expressing cells are
cultured in a medium containing recombinant insulin and a
human plasma protein solution. Sucrose is used as a
stabiliser rather than human albumin. The purification
process includes two viral inactivation steps by
solvent/detergent treatment and high salt treatment.
3rd
generation
rFVIII
Advate® (Baxter)c
Factor VIII genes are inserted into Chinese hamster ovary
cells. The rFVIII is purified using a series of chromatography
columns, using an immunoaffinity chromatography step with
a monoclonal antibody to isolate the rFVIII from the medium.
This is further purified with a viral inactivation solvent
detergent. No animal or human products are used in the cell
culture or purification processes.
3rd
generation
rFIX
BeneFIX® (Wyeth
Pharmaceuticals)
Factor IX genes are inserted into Chinese hamster ovary
cells. The secreted FIX is purified with four chromatographic
steps and three (nanofiltration, ultrafiltration and diafiltration)
membrane based filtration steps, the final of which is a viral
retention filtration step. Human albumin and immunoaffinity
chromatography are not used.
The treatment of
choice for the control
and prevention of
haemorrhagic
episodes and for
routine and surgical
prophylaxis in
patients with
haemophilia B.
2nd
generation
rFVIIa
NovoSeven® (Novo
Nordisk)
Baby hamster kidney cells are genetically transformed to
produce factor VII as a single-chain glycoprotein (406 amino
acids, 50kDa). This is purified by ion exchange and
immunoaffinity chromatography using murine monoclonal
antibodies. The rFVII is converted to the two-chain activated
form, and freeze dried. Trace amounts of hamster proteins,
mouse proteins, bovine IgG and other bovine products are
included.
For the control and
prevention of
haemorrhagic
episodes and for
routine and surgical
prophylaxis in
patients with
haemophilia A or B
and inhibitors to FVIII
or FIX. May also be
used for patients with
FVII deficiency.
modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000 and information from the Therapeutic Goods
Administration (accessed 2004); bManufacturers are current as at January 2005, but are subject to change; cAdvate® (Baxter) is currently
registered as an Orphan drug by the Therapeutic Goods Administration; FVII= factor VII; FVIII factor VIII; FIX= factor IX; IgG=Immunoglobulin
G; rFVIIa= activated recombinant factor VII; rFVIII= recombinant factor VIII; rFIX= recombinant factor IX; vWF= von Willebrand factor
a
Use of recombinant and plasma-derived Factor VIII and IX
33
Prothrombin complex concentrates (PCCs)
Activated prothrombin complex concentrates, which directly activate prothrombin by
stimulation of activated factor X (Xa), are useful in treating patients with inhibitors to
factor VIII or IX (Kingdon & Lundblad 2002) (Table 7).
Table 7.
Prothrombin complex concentrates (PCCs)*
Type of
product
Product
(Manufacturer)
Manufacture of product
Therapeutic use of
product
PCCs
Prothrombinex®-HT
(CSL)
This concentrate of factors II, IX and X is prepared from
cryoprecipitate supernatant and purified by ion exchange
chromatography. It is lyophilised and then heat-treated
for 72 hours at 80°C for viral inactivation.
Prothrombinex® contains approximately 500 units each
of factors II, IX and X.
Traditionally used for
treatment in haemophilia
B, but now replaced by
BeneFIX® and
MonoFIX®. Treatment of
choice for FII or FX
deficiencies.
aPCCs
FEIBA® (Baxter)
This activated prothrombin complex concentrate is
prepared from cryoprecipitate supernatant which then
generates FEIBA. This is purified with adsorption and
filtration and then vapour heated for 10 hours at 60°C
then 80°C for 1 hour before being lyophilised.
For the control and
prevention of
haemorrhagic episodes
and for routine and
surgical prophylaxis in
patients with haemophilia
A or B and inhibitors to
FVIII or FIX.
* modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000 ; aPCC= activated prothrombin complex
concentrates; FEIBA= factor eight inhibitor bypassing agent; FII= factor II; FVIII= factor VIII; FIX= factor IX; FX= factor X; PCC= prothrombin
complex concentrates
Coagulation factor concentrates for less common disorders
Haemophilia A and B and von Willebrand disease account for the majority of inherited
bleeding disorders. The remainder include deficiencies of fibrinogen, prothrombin, and
deficiencies of factor V, combined V/VIII, VII, X, XI, and XIII. While treatments for
haemophilia have improved over the last 15 years, treatments for more rare deficiencies
traditionally have used fresh frozen plasma and cryoprecipitate. For deficiencies in factor
V, fresh frozen plasma remains the only treatment option since cryoprecipitate does not
contain any factor V, and factor V concentrate is not available. However, for other
deficiencies, plasma-derived concentrates are now available. Since factor XI has a half-life
of two to three days and has been associated with thrombosis, factor XI concentrates are
generally used in low doses for surgical procedures and control of bleeding associated
with trauma (Kasper 2004). People with a deficiency in factor XIII are at risk of
intracranial bleeding and pregnant women have a high risk of miscarriage. Factor XIII
has a long half-life of 10 to 20 days and monthly prophylactic infusions with factor XIII
concentrates may be a convenient and effective way of preventing spontaneous bleeding.
Due to the small demand for concentrates for rare bleeding disorders, it is unlikely that
recombinant products will be developed or become available in the near future.
Therefore, plasma-derived factor concentrates are likely to remain the treatment of
choice for these conditions until gene therapy is available (Di Paola et al. 2001). Table 8
lists the coagulation factor concentrates that are available for the less common
coagulation disorders.
34
Use of recombinant and plasma-derived Factor VIII and IX
Table 8.
Coagulation factor concentrates for less common disorders*
Type of
product
Product
(Manufacturer)
Manufacture of product
Therapeutic use of
product
Fibrinogen
Fibrinogen (Immuno)
Cryoprecipitate supernatant is treated with DEAEsephadex to produce fibrinogen, which is then virally
inactivated with vapour heating.
For use in patients with
fibrinogen deficiency.
Haemocomplettan P
(ZLB Behring)
The glycine supernatant from the intermediate-purity
FVIII process produces fibrinogen which is pasteurised
for 10 hours at 60°C.
Factor VII (Baxter)
This high purity concentrate is manufactured using twostep vapour heat inactivation for 10 hours at 60°C then
80°C for 1 hour, both under excess barometric pressure.
A PCR test for HIV, HBV and HCV is carried out.
Factor VII (Bio
Products Laboratory)
This high purity concentrate is manufactured with a
dedicated viral inactivation procedure. Plasma is sourced
from FDA approved USA sites.
Facteur VII (LFB)
Fractionated by DEAE adsorption and anion exchange
chromatography. Virally inactivated by TNBP/polysorbate
80; no albumin added (Kasper 2004)
Factor XI (Bio
Products Laboratory)
This high-purity concentrate is dry heated at 80ºC for 72
hours to inactivate viruses. Since 1993, this product has
included heparin and antithrombin to minimise
thrombogenicity (Bolton-Maggs 2000).
Hemoleven (LFB)
This product, which is similar to the BPL factor XI, is
manufactured in France. It contains smaller amounts of
heparin and antithrombin than in the BPL product and
viral inactivation is by solvent/detergent and
nanofiltration.
Factor XIII (Bio
Products Laboratory)
This high purity concentrate is manufactured with a
dedicated viral inactivation procedure. Plasma is sourced
from FDA approved USA sites.
Fibrogammin® P (ZLB
Behring)
This FXIII concentrate is prepared from cryoprecipitate
supernatant and then purified by ion exchange
chromatography. It is then pasteurised for 10 hours at
60°C, and albumin is added as stabiliser.
pdFVII
pdFXI
pdFXIII
To be used for FVII
deficiency at a dose of 5–
10 IU/kg.
For use in FXI deficient
people to treat excessive
bleeding due to trauma or
surgery. Due to the long
half-life (2–3 days), the
dosage required for
surgery is low. Restraint
in dosage is
recommended due to
possible thrombogenicity,
particularly in the elderly
and/or those with preexisting cardiovascular
disease. Factor XI should
not be given at doses
exceeding 30 U/kg
(product information).
For use in FXIII deficient
patients. Patients should
be monitored for
development of inhibitors
to FXIII under clinical
observation and
laboratory tests. Patients
on a low sodium diet
should be closely
monitored as FXIII
concentrate contains
sodium chloride. May be
used every 21 days in
individuals with a history
of spontaneous abortions
(Di Paola et al 2001)
* modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000 ; DEAE= diethylaminoethyl; FDA= Food and Drug
Administration (USA); FVII= factor FVII; HBV= hepatitis B; HCV= hepatitis C; HIV= human immuno-deficiency virus; LFB= Laboratoire Francais
du Fractionnement et des Biotechnologies; PCR= polymerase chain reaction; pdFVII= plasma-derived factor VII; pdFXI= plasma-derived factor
XI; pdFXIII= plasma-derived factor XIII; TNBP= tri(n-butyl)phosphate
Gene therapy
Gene therapy involves insertion of the factor VIII/IX gene or an active derivation into
the cells of a patient, allowing expression and secretion of factor proteins into the blood
stream (Kingdon & Lundblad 2002). Although research in gene transfer is progressing
Use of recombinant and plasma-derived Factor VIII and IX
35
(Powell et al. 2003; Roth et al. 2001b), this field is still in its infancy and results have been
limited. Therefore, factor replacement therapy remains the mainstay of haemophilia care.
36
Use of recombinant and plasma-derived Factor VIII and IX
Adjunctive haemostatic agents
Due to the risks associated with viral transmission and the high cost of factor
concentrates, a range of haemostatic agents (Table 9) have been trialled for use in
patients with haemophilia A, B or von Willebrand disease (vWD). While the routine use
of viral inactivation techniques and the development of recombinant products has
alleviated most of the anxieties about the safety of factor concentrates, they are still
expensive and other products may be considered if necessary (Saulnier et al. 1994;
Warrier & Lusher 1983).
Table 9.
Adjunctive haemostatic agents*
Type of
product
Product
(Manufacturer)
Description of product
Therapeutic use of product
DDAVP
Desmopressin acetate
Octostim (Fisons
Pharmaceuticals),
Minirin (Ferring)
Desmopressin acetate is a synthetic
analogue of the anti-diuretic hormone
8-D-arginine vasopressin.
DDAVP is for use in mild/moderate
haemophilia A or mild vWD.
Contraindicated in those with
arteriovascular disease, those under two
years of age, and should be used with
caution in the elderly or in pregnant
women.
Fibrin
sealant
Tisseel Duo (Baxter)
This fibrin sealant contains human
fibrinogen, fibronectin, FXIII and
plasminogen. The product is double
steam heat-treated and PCR tested
for HIV, HBV and HCV. The end
product is deep-frozen.
To be used locally as an adjunct to
haemostasis during surgical procedures,
when control of bleeding by conventional
surgical techniques is ineffective or
impractical.
Tranexamic
acid
Cyklokapron (Pfizer)
Tranexamic acid inhibits activation of
plasminogen and plasmin, and is
available in both suspension and
tablet form.
Particularly useful for treating bleeding
from gastrointestinal tract, menorrhagia,
open wounds, dental surgery and in
conjunction with DDAVP. Treatment of
choice for patients with FXI deficiency
for mild bleeding episodes.
*modified from Haemophilia Foundation Australia Medical Advisory Panel guidelines 2000; DDAVP= 1-deamino-8-D-arginine vasopressin;
FXI= factor XI; HBV= hepatitis B; HCV= hepatitis C; HIV= human immuno-deficiency virus; PCR= polymerase chain reaction; vWD= von
Willebrand Disease
Desmopressin (1-deamino-D-arginine vasopressin, DDAVP)
DDAVP, which is a synthetic analogue of the natural anti-diuretic hormone arginine
vasopressin, induces the release of factor VIII and von Willebrand factor into the
bloodstream. Plasma levels peak 30–60 minutes after administration and the half-life is
comparable to levels found after administration of factor concentrates (Association of
Hemophilia Clinic Directors of Canada 1995b).
Previous Australian consensus-based guidelines
DDAVP should be considered for all patients with mild/moderate haemophilia A or mild vWD. DDAVP is
generally administered intravenously at a dose of 0.3 mcg/kg diluted in 50 mL of 0.9% saline and
infused over at least 30 minutes. Efficacy should be demonstrated irrespective of the route employed,
by measuring FVIII/vWF. DDAVP should be used with caution in elderly individuals, pregnant women
and avoided in those with evidence of arteriovascular disease and in children younger than 2 years of
age. (see Appendix G for full details) (Haemophilia Foundation Australia Medical Advisory
Panel 2000).
Use of recombinant and plasma-derived Factor VIII and IX
37
Three randomised controlled trials and one non-randomised controlled trial (Level II and
III-2 evidence) examining effectiveness and/or safety of DDAVP were included in the
systematic review for assessment. The other included studies on DDAVP were case
series (Level IV evidence) (Table 10).
Effectiveness of DDAVP
DDAVP is generally available in Australia in two strengths in both intravenous and
subcutaneous form. The different modes of administration are comparable at the same
0.3 µg/kg dose (Level II evidence) (Mannucci et al. 1987). One study compared the
effectiveness and safety of a 0.3 µg/kg dose with a 0.4 µg/kg dose and concluded that
the 0.3 µg/kg dose had the same efficacy as the 0.4 µg/kg dose (Level IV evidence)
(Mariana et al. 1984).
One good quality case series had one patient who was given DDAVP by mistake during
labour. It was found that DDAVP appeared to stop the process of labour when
administered when the cervix was fully dilated (Schulman et al. 1991). While it is unclear
whether it was the DDAVP or a natural occurrence in a primipara, similar observations
have been made in case reports (below the level of evidence examined in this review).
Consequently, if DDAVP is to be given to prevent blood loss in labour, it should be
given postpartum to prevent interference in labour, at least until stronger evidence
refutes the recommendation (expert opinion) (Schulman et al. 1991).
One good quality randomised crossover trial (Level II evidence) showed that a DDAVP
nasal spray alleviated menorrhagia in women with inherited bleeding disorders (subjective
assessment). However, statistically the effect was no greater than that of placebo due to
the small sample size (statistically underpowered) (Level II) (Kadir et al. 2002). In
Australia, the intranasal spray is available only through individual importation.
The best evidence available on the effectiveness of DDAVP showed that in multitransfused patients with severe vWD (bleeding time over 30 minutes) receiving
cryoprecipitate for joint or muscle haemorrhages, DDAVP assisted in further reducing
bleeding time to within the normal range (upper limit seven minutes), showing a
statistically significant improvement in haemostatic control and a clinical benefitd
compared to saline placebo (Level II evidence) (Cattaneo et al. 1989).
Lower levels of evidence have shown that DDAVP alone is unsuitable for use in severe
vWD, since only 18–27% of patients with severe Type 1 and Type 2 vWD met the
criteria for response to DDAVP (Level IV) (Federici et al. 2004). Although the criteria
for minimum response and definition of a positive haemostatic effect differed slightly
between studies, generally they referred to the increase of factor concentrates and
reduction of bleeding time to within the normal rangee.
Clinical importance of statistically significant effects was ranked according to the checklist provided in
Appendix H. More than 20% improvement in haemostasis—vs baseline—indicated a clinical benefit.
d
e The normal range is defined according to the results found in individual laboratories. Normal factor
concentrate levels found by Nolan et al. 2000 were FVIII:C=0.5-1.5 IU/dl, vWF:Ag=0.5-1.5 IU/dl, and
vWF:Ac=0.5-1.5 IU/dl. Normal bleeding time as defined by Federici et al. 2004 was 12 minutes or less.
38
Use of recombinant and plasma-derived Factor VIII and IX
DDAVP was clinically effective in the majority of mild and moderate vWD and
haemophilia A patients. However, a significant number of patients did not meet the
minimum response criteria (Level IV evidence) (Nolan et al. 2000; Federici et al. 2004;
Schulman et al. 1991). Response to DDAVP depends on both the severityf and the
subtype of disease, and the mode of administration. While the sample size of study
participants within particular subgroups, other than for Type 1 vWD, were small, the
available evidence suggests that Type 1 and mild Type 2 vWD patients are likely to
respond to DDAVP. In contrast, severe Type 2 and Type 3 vWD are unlikely to respond
measurably unless DDAVP is used as an adjunctive agent after factor concentrates have
reduced bleeding (Schulman et al. 1991; Federici et al. 2004; Nolan et al. 2000; Cattaneo
et al. 1989). Thus, since approximately 20–30% of patients are non-responsive to
DDAVP, it is suggested that a test dose should be administered initially to establish
efficacy.
When patients repeatedly receive DDAVP in a short space of time, they may become
less responsive or unresponsive to its effects (tachyphylaxis). A low level study by
Mannucci et al. 1992 found that when DDAVP was administered on four consecutive
days, the response to DDAVP on the second day was 30% lower than on the first and
continued to decline over the subsequent three days. The development of tachyphylaxis
limits the usefulness of DDAVP over a long period of time (Level IV evidence).
DDAVP has also been effective in preventing, stopping or reducing excessive bleeding
in dental extractions and surgery. Data from studies using DDAVP in surgical/dental
procedures are presented in the Surgical and Dental Procedures section.
f Severe vWD was defined by participating centres as a lifelong history of bleeding, including ≥ 2 episodes
requiring replacement therapy and at least one of the following laboratory abnormalities: bleeding time >
15 minutes; vWF:RCo < 10 IU/dL; or FVIII:C < 20 IU/dL (Federici et al. 2004).
Use of recombinant and plasma-derived Factor VIII and IX
39
Table 10.
Study
(Kadir et al.
2002)
Effectiveness of DDAVP for haemostatic control
Level and
quality of
evidence
Population
Level II:
randomised
crossover trial
39 women
with mildmoderate
vWD,
heterozygote
FXIdeficiency,
and carriers
of
haemophilia,
aged 18–60
Quality score:
24/27
External validity
uncertain
Bias minimised
Confounding
avoided
(Mannucci et al.
1987)
Level II:
randomised
crossover trial
Quality score:
23/27
External validity
uncertain
Bias minimised
Level II:
randomised
crossover trial
Quality score:
20/27
Clinical
importancec:
1/4
External validity
uncertain
PBAC Scores
14 mild and
moderate
haemophilia
A (median
FVIII:C level
7 U/dL, range
2-31)
Test dose
Quality score:
17/27
External validity
uncertain
Bias possible
Confounding
possible
40
140
148
Mean difference= -8
No statistically significant difference,
p=0.51
Intravenous
DDAVP (0.3
µg/kg)
mean±SD
Subcutaneous
DDAVP (0.3 µg/kg)
mean±SD
Peak FVIII:C (pg/mL)
2363d
582±158
Half-life of FVIII:C (hr)
4.4±1.1
4.7±1.6
Significant difference in peak FVIII:C (p<0.02)
10 PTPs with
severe vWD,
(type not
specified)
with
prolonged
bleeding time
(>30 minutes)
Cryoprecipitate +
DDAVP (0.3 µg/kg)
mean±SD
Cryoprecipitate +
saline
mean±SD
Post cryo=14±2
Post DDAVP=9±2
Post cryo=15±3
Post saline=15±3
p<0.01
NS
Post cryo=75±7
Post DDAVP=69±6
Post cryo=92±9
Post saline=83±1
NS
NS
Post cryo=64±7
Post DDAVP=64±7
Post cryo=76±8
Post saline=63±7
NS
NS
Within subjects
Intravenous
DDAVP (0.3 µg/kg)
mean±SD
Subcutaneous
DDAVP (0.3 µg/kg)
mean±SD
FVIII:C (IU/dl)
24.9±12.4
Range= 9.5-50.0
25.5±12.4
Range= 12.8-48.0
Ratio post/pre
3.7±1.4
Range= 1.7-5.9
3.1±1.4
Range= 1.7-5.1
Bleeding time
(minutes)
vWF:Ag (IU/dL)
Test dose
vWF:RCo (IU/dL)
Confounding
avoided
Level III-2: non
randomised
controlled trial
Placebo
Non-significant difference in half life of FVIII:C
Bias minimised
(De Sio et al.
1985)
DDAVP (300µg)
Prophylaxis
for
menorrhagia
Confounding
avoided
(Cattaneo et al.
1989)
Effectiveness
26 patients,
21 mild
haemophilia
A, 5
moderate
haemophilia
A
8 patients
had both
treatments
Test dose
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 10. (cont.)
(Schulman et al.
1991)
Level IV: before
and after study
Quality score:
3/3
Measurement
bias minimised
Selection bias
minimised
Follow-up
adequate
370 patients
with disorders
of primary
haemostasis.
133 vWD,
237 platelet
function
defects, aged
3–80 years
Test dose
Uncontrolled
(Federici et al.
2004)
Level IV: before
and after study
Quality score:
3/3
Measurement
bias minimised
Selection bias
minimised
(Nolan et al.
2000)
66 patients
with severeb
Type 1 or 2
vWD, BT
more than 15
minutes,
aged 12–65
years
Follow-up
adequate
Exclusions:
Type 2B and
type 3 vWD
Uncontrolled
Test dose
Level IV: before
and after study
133 patients
with bleeding
disorders. 91
Type 1 vWD,
20 mild
haemophilia
A, 22 platelet
function
disorders
Quality score:
3/3
Measurement
bias minimised
Selection bias
minimised
Follow-up
adequate
No. responsivea to DDAVP (0.2–0.3 µg/kg)
Total vWD
120/133 (90%)
Type 1 (mild) vWD
110/113 (97%)
Type 1 (moderate) vWD
6/10 (60%)
Type 2A vWD
3/3
Type 2B vWD
1/4
Type 3 (severe) vWD
0/3
Platelet function disorder
202/237 (85%)
No.
responsivea
to DDAVP (0.3 µg/kg)
Type 1 vWD
7/26 (26.9%)
Type 2A vWD
1/15 (0.1%)
Type 2M vWD
3/21 (14.2%)
Type 2N vWD
3/4 (75.0%)
No. responsivea to DDAVP (0.3 µg/kg)
Type 1 vWD
82/91 (90.1%)
Mild haemophilia A
15/20 (75.0%)
Platelet function disorders
15/22 (68.2%)
Test dose
Uncontrolled
(Mannucci et al.
1992)
Level IV: before
and after study
Quality score:
2/3
Measurement
bias minimised
Selection bias
possible
Follow-up
adequate
Uncontrolled
37 patients,
22 with mild
haemophilia
A, 15 with
mild Type 1
vWD, aged
16–63 years
Test doses
DDAVP (0.3 µg/kg)
Day 1
Day 2
Day 3
Day 4
Haemophilia
FVIII:C
post/pre‡
3.0±0.7
2.0±2.5*
2.0±0.6*
1.9±0.5*
vWD
FVIII:C
post/pre‡
4.3±1.9
3.4±2.7*
3.5±2.8*
2.8±1.2*
vWF:Ag
post/pre‡
4.8±2.4
3.1±1.8*
3.4±1.8*
3.3±2.3*
RCo
post/pre‡
5.1±3.0
4.3±3.3*
3.8±2.4*
3.5±1.8*
BT
post/pre‡
0.6±0.2
0.6±0.2
0.7±0.2
0.7±0.2
‡ Data
are expressed as a post/pre DDAVP ratio.
*significantly less response than Day 1 (p<0.001)
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
41
Table 10 (cont.)
Study
(Warrier &
Lusher 1983)
Level and
quality of
evidence
Level IV: before
and after study
Quality score:
2/3
Measurement
bias minimised
Population
3 vWD
patients
3 cases of
menorrhagia
Effectiveness
DDAVP via intranasal drip (2–4 µg/kg) or intravenous (0.2–0.3
µg/kg)
All reported decreased menstrual bleeding within 24 hours after
receiving DDAVP
Selection bias
possible
Follow-up
adequate
Uncontrolled
(Leissinger et al.
2001)
Level IV: before
and after study
Quality score:
1/3
172 Type 1
vWD
patients:
DDAVP (150 µg) intranasal
Mild type 1 vWD:
135 mild
211/254 (83%) rated haemostasis ‘Excellent’; 35/254 (14%) rated
‘Good’
Measurement
bias minimised
37 moderate
Moderate type 1 vWD:
Selection bias
possible
254 bleeding
episodes
Follow-up
inadequately
stated
55/78 (71%) rated haemostasis ‘Excellent’, 14/78 ‘Good’.
Uncontrolled
(Mariana et al.
1984)
Level IV: before
and after study
Quality score:
1/3
Measurement
bias possible
Selection bias
likely
Follow-up
adequate
5 vWD
patients
(Types 1 and
2)
DDAVP (0.3–0.4 µg/kg) intravenous + tranexamic acid
Bleeding or pain ceased promptly in 6/7 episodes, 1 progressive
reduction in bleeding.
7
spontaneous
bleeding
episodes
Uncontrolled
the definition of responsiveness in the normal range varies across laboratories. Normal factor concentrate levels found by Nolan et al. 2000
were FVIII:C=0.5-1.5 IU/dl, vWF:FVIIIB, and vWF:Ag=0.5-1.5 IU/dl; Normal bleeding time was defined by Federici et al. 2004 as 12 minutes or
less; b Severe vWD was defined by participating centres as a lifelong history of bleeding, including ≥ 2 episodes requiring replacement therapy
and at least one of the following laboratory abnormalities: bleeding time > 15 minutes; vWF:RCo < 10 IU/dL; or FVIII:C < 20 IU/dL (Federici et
al, 2004); c Rank scores for the clinical importance of the benefit/harm (1/4 ranked highest and 4/4 ranked lowest); d Information on population
source not provided; BT= bleeding time, in minutes; cryo= cryoprecipitate; FVIII:C= factor VIII coagulant activity; FXI= factor XI; PBAC=
Pictorial Blood Assessment Chart, a commonly used tool to measure menstrual loss where a score of ≥100 is equivalent to more than 80 mL of
blood loss (definition of menorrhagia); PTPs= previously treated patients; RCo= ristocetin cofactor; SD= standard deviations; vWD=von
Willebrand Disease; vWF:Ag= von Willebrand factor antigen; d Standard deviation provided by the authors was not meaningful
a
Safety of DDAVP
The majority of studies on the safety of DDAVP were too small to be able to identify
uncommon side effects (Table 11). The best evidence addressing the side effects of
DDAVP found that while there were numerous adverse events related to the use of a
DDAVP intranasal spray to reduce menorrhagia in women with vWD and other rare
bleeding disorders, there were a similar number of adverse events related to the use of
placebo (Level II evidence) (Kadir et al. 2002).
42
Use of recombinant and plasma-derived Factor VIII and IX
With the exception of transient headaches and/or mild facial flushing, the majority of
papers reported few side effects from DDAVP (Rose & Aledort 1991; Warrier & Lusher
1983; Chuansumrit et al. 1993; Rodeghiero et al. 1996; Dewald et al. 1980; Ghirardini et
al. 1988; Mariana et al. 1984). In some cases, more severe, but less common, side effects
related to DDAVP included dizziness, nausea, vomiting, asthenia, and side effects
associated with excess fluid, such as oedema and hyponatraemia (Schulman et al. 1991;
Ghirardini et al. 1987; Dunn et al. 2000; Gill et al. 2002; de la Fuente et al. 1985;
Leissinger et al. 2001).
DDAVP has an anti-diuretic effect, which means patients must restrict their fluid intake
to avoid the adverse events associated with low levels of sodium in the blood. Fluid
restriction may be based on the following calculation: 100 mL/kg for the first 10 kg of
body weight + 50 mL/kg for the second 10 kg of body weight + 20 mL for each
additional kilogram of body weight (Gill et al. 2002). A simpler restriction is the limit of
1.5-2 L of fluid per day for adults (Kadir et al. 2002). For the most part, side effects
associated with DDAVP are minimised when patients adhere to fluid restrictions (Level
IV evidence) (Dunn et al. 2000). Alternatively, close observation of sodium and symptom
status may be adequate (expert opinion) (Palmer et al. 2003). If DDAVP is given more
than once in 24 hours, predose monitoring of electrolyte concentrations is recommended
(JBC FVIII/FIX working party & National Blood Authority 2004).
No studies included in this systematic review evaluated the safety of DDAVP in children
less than two years of age. Therefore, any such recommendation would be based on
expert opinion. Previous recommendations have suggested DDAVP should be avoided
in those younger than two years of age due to risk of fluid overload (Haemophilia
Foundation Australia Medical Advisory Panel 2000).
Use of recombinant and plasma-derived Factor VIII and IX
43
Table 11.
Adverse events associated with use of DDAVP
Study
Level and quality of
evidence
(Kadir et al. 2002)
Level II: randomised
crossover trial
Quality score: 24/27
External validity
uncertaina
Bias minimised
(De Sio et al. 1985)
Population
39 women with
menorrhagia. Mildmoderate vWD,
heterozygote FXIdeficiency, and
carriers of
haemophilia
Adverse events to DDAVP
24/29 (83%) adverse events.
Headache, facial flushing, weight gain
DDAVP (300µg)
Placebo
Total
18/26 (69%)
15/29 (52%)
Headache
23%
24%
12%
0%
Confounding unlikely
Prophylaxis for
menorrhagia
Weight
gain
Level III-2: non
randomised
controlled trial
26 patients, 21 mild
haemophilia A, 5
moderate
haemophilia A
DDAVP (0.3 µg/kg)
intravenous
DDAVP (0.3 µg/kg)
subcutaneous
Moderate increase in
pulse rate and/or blood
pressure, frequently
associated with mild
facial flushing
Immediate swelling
caused by injection,
disappeared within a few
minutes
Quality score: 17/27
Test dose
External validity
uncertain
Bias possible
Confounding
possible
(Nolan et al. 2000)
Level IV: prospective
case series
Quality score: 3/3
Measurement bias
minimised
Selection bias
minimised
133 patients with
bleeding disorders.
91 Type 1 vWD, 20
mild haemophilia A,
22 platelet function
disorders
No other adverse events
reported
DDAVP (0.3 µg/kg) intravenous infusion
2/133 minor side effects:
Facial flushing and palpitations—resolved by
decreasing infusion rate
Test dose
Follow-up adequate
Uncontrolled
(Schulman et al.
1991)
Level IV: case series
Quality score: 3/3
Measurement bias
minimised
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Rodeghiero et al.
1996)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
370 patients with
disorders of primary
haemostasis. 133
vWD, 237 platelet
function defects,
aged 3–80 years
127 patients given
DDAVP on 173
occasions, either
prophylaxis for
surgery or treatment
for bleeding
79/169 patients, 43
vWD, 36 haemophilia
A
219 bleeding
episodes—including
menorrhagia, tooth
extraction, epistaxis,
muscle haematoma
and haemarthrosis
DDAVP (0.2–0.3 µg/kg, route of administration
not specified)
Two patients had side effects after surgery, relating
to fluid retention. Mild side effects such as nausea,
dizziness, or moderate fatigue in patients undergoing
general anaesthesia could not solely be attributed to
desmopressin
DDAVP (20 µg for >70 kg or 40 µg for <70 kg)
subcutaneous in home treatment
30% of patients reported mild flushing, with or
without headache. One patient had mild chest pain
unlikely to be related to treatment
(cont.)
44
Use of recombinant and plasma-derived Factor VIII and IX
Table 11 (cont.)
Level IV: case series
(de la Fuente et al.
1985)
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
(Dunn et al. 2000)
Level IV: case series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Warrier & Lusher
1983)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
(Ghirardini et al.
1987)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias likely
68 patients, 32 mild
and 8 moderate–
severe haemophilia
A, 13 Type 1 vWD, 7
Type 2A vWD, 1
Type 2B vWD, aged
2–66 years
26 test doses, 17
bleeding episodes,
12 dental extractions,
24 surgical
interventions
40 patients, 24
Types 1 and 2 vWD,
16 mild–moderate
haemophilia A and
symptomatic
haemophilia A
carriers aged 5–58
years
Test dose
38 patients, 31 vWD,
7 mild–moderate
haemophilia A and 3
normal subjects
DDAVP (0.3 µg/kg) intravenous
24/68 adverse events:
A ‘warm feeling’ or mild facial erythema. Other
possible side effects may have been due to preexisting medical problems, ie headaches (n=4),
fatigue (n=2), oedema with transient rash (n=1). One
patient was hyponatraemic and had a grand mal
seizure after vomiting
DDAVP intranasally, 150 µg if under 50k g,
300 µg if over 50 kg
27/40 side effects. 75% of vWD patients and 56% of
haemophilia A patients. 78% had no or mild
reactions (facial flushing, transient headache,
burning/itching eyes) while 23% had moderate to
severe adverse events (nausea, fatigue, moderatesevere headache, abdominal cramping, weight gain,
vomiting, vertigo, ataxia, weakness)
DDAVP via intranasal drip (2–4 µg/kg) or
intravenous (0.2–0.3 µg/kg)
No side effects
8 surgical events, 3
cases of
menorrhagia, 4
patients with
bleeding episodes
25 patients, 23 mild
haemophilia A, 2
moderate
haemophilia A
Test dose
DDAVP (0.3 µg/kg) subcutaneous
5/25 adverse events:
Modest facial flushing, transient headache, brief
nausea
Follow-up adequate
Uncontrolled
(Gill et al. 2002)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
25 patients, 9
haemophilia A, 16
vWD,
Test dose
Selection bias
possible
DDAVP intranasal (150 µg for under 50 kg)
12/25 (48%) 16 adverse events.
Headache, congestion, fatigue, flushing, difficulty
breathing, insomnia, generalised leg aches and stiff
neck
Follow-up adequate
Uncontrolled
(Ghirardini et al.
1988)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
24 patients, 16 mild
haemophilia A, 2
moderate
haemophilia A, 6
Type 1 vWD
patients,
10 bleeding
episodes, 6 dental
extractions, 11
surgical interventions
Use of recombinant and plasma-derived Factor VIII and IX
DDAVP subcutaneous (0.3 µg/kg)
A few reports of mild and transient facial flushing
(cont.)
45
Table 11 (cont.)
Study
Level and quality of
evidence
Population
(Rose & Aledort
1991)
Level IV: case series
22 patients, 11 vWD,
8 mild haemophilia
A, 3 symptomatic
carriers of
haemophilia A
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Test doses
Adverse events to DDAVP
DDAVP (300 µg for adults, 150 µg for childrenunspecified age) intranasally
Approximately 50% nasal spray administrations
resulted in facial flushing and/or transient headache.
No other adverse events
Follow-up adequate
Uncontrolled
(Chuansumrit et al.
1993)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
(Dewald et al. 1980)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
22 patients, 13
haemophilia A, 1
Type 1 vWD, 8 with
inherited or acquired
platelet dysfunction,
aged 2–26 years
DDAVP (0.3–0.4 µg/kg) intravenous
No adverse events
31 administration
episodes, either
treatment of bleeding
episode or test dose
5 patients, 4
haemophilia A, 1
vWD
DDAVP (0.4 µg/kg) intranasal
No side effects
10 test doses
Selection bias
possible
Follow-up adequate
Uncontrolled
(Mariana et al. 1984)
Level IV: case series
Quality score: 1/3
Measurement bias
possible
Selection bias likely
Follow-up adequate
Uncontrolled
(Leissinger et al.
2001)
Level IV: case series
Quality score: 1/3
Measurement bias
minimised
Selection bias
possible
Follow-up
inadequately stated
Uncontrolled
a
43 patients, 21 mild
haemophilia A, 2
moderate
haemophilia A, 20
vWD (Types 1 and 2)
DDAVP (0.3 µg/kg)
intravenous
52 infusions, no side
effects
DDAVP (0.4 µg/kg)
intravenous
13 infusions, no side
effects
29 dental extractions,
12 surgical
procedures, 12
spontaneous
bleeding episodes
333 patients
enrolled. 124 mild
haemophilia A, 135
mild Type 1 vWD, 37
moderate Type 1
vWD, 23 haemophilia
carriers, 14 other
bleeding disorders,
aged 5–64 years
45% of doses treated
748 episodes of
bleeding, 19%
prophylaxis, 35% for
menorrhagia
DDAVP (150 µg) intranasal
Overall 80 patients reported 272 adverse events. 80
had headaches, 59 had vasodilation/flushing, 33 had
nausea, 23 had dizziness, 14 had asthenia, 8
vomited, 6 had peripheral oedema, and 49 had less
common adverse events
No. of adverse events/ no. of patients
Mild Haemophilia A
80/124
Mild Type 1 vWD
117/135
Moderate Type 1 vWD
37/37
Haemophilia A carrier
24/23
Other bleeding disorders
14/14
Information on population source not provided; DDAVP, 1-deamino-D-arginine vasopressin; vWD= von Willebrand Disease
46
Use of recombinant and plasma-derived Factor VIII and IX
Evidence-based clinical practice guidelines
On the basis of the available research, there are several modifications to the existing recommendations:
•
DDAVP may be considered for all patients with mild/moderate haemophilia A or mild vWD. DDAVP
is generally administered intravenously at a dose of 0.3 µg/kg diluted in 50 mL of 0.9% saline and
infused over at least 30 minutes. Subcutaneous administration of DDAVP is comparable (Level II
evidence). Efficacy should be demonstrated with a test dose irrespective of the route employed, by
measuring FVIII/vWF. DDAVP should be used with caution during pregnancy and in elderly
individuals (Level IV evidence). It is not recommended in those with evidence of arteriovascular
disease and in young children (<2 years) (expert opinion).
•
DDAVP is not recommended during labour. The evidence suggests that it is clinically effective in
reducing bleeding when administered immediately postpartum (Level IV evidence).
•
DDAVP may be considered for use prior to dental extractions or surgery in patients who respond to
DDAVP (Level IV evidence).
•
DDAVP should not be used as first treatment for patients with severe vWD. Type 1 and Type 2A
vWD patients are deemed likely to respond, whereas Type 2B and Type 3 vWD are deemed
unlikely to respond (Level IV evidence). However, DDAVP may assist as an adjunct to factor
replacement therapy (Level II evidence).
•
Caution should be taken to restrict fluid intake during DDAVP treatment to prevent fluid overload,
leading to hyponatraemia (Level IV evidence). Daily measurement of serum sodium levels may be
taken (expert opinion).
•
DDAVP may be administered once during every 24 hours. If given for more than three consecutive
days, repeated doses may lead to tachyphylaxis (Level IV evidence). If DDAVP is given more than
once in 24 hours, predose monitoring of electrolyte concentrations is recommended (expert
opinion).
Use of recombinant and plasma-derived Factor VIII and IX
47
Tranexamic acid
Previous Australian consensus-based guidelines
•
Tranexamic acid is an antifibrinolytic agent that competitively inhibits the activation of plasminogen
to plasmin and is available in an intravenous or oral preparation. Tranexamic acid is particularly
useful for bleeding from the gastrointestinal tract, menorrhagia, open wounds, dental surgery and
in conjunction with DDAVP.
•
The recommended intravenous dose is 10 mg/kg 2–3 times daily and the oral dose 25 mg/kg 2-3
times daily. Tranexamic acid is contraindicated in patients with thromboembolic disease and
should be avoided in patients with haematuria. It should not be used with Factor Eight Inhibitory
Bypass Agents (FEIBA) or other prothrombin complex concentrates. Tranexamic acid can be used
in conjunction with recombinant human coagulant factor VIIa bypassing agent.
See Appendix G for full details (Haemophilia Foundation Australia Medical Advisory Panel 2000)
Tranexamic acid is usually used in conjunction with other treatments, such as DDAVP,
fibrin glue or factor replacement. Very few studies have evaluated the effects of
tranexamic acid alone in individuals with coagulation disorders. One average quality
randomised controlled trial (Level II evidence) investigated the effectiveness, and three
case series (Level IV evidence) reported on the safety, of tranexamic acid (Table 12 and
Table 13). Tranexamic acid was found to reduce the amount of factor concentrates
needed in a group of patients with severe haemophilia A, but this difference was not
statistically significant (Schiavoni et al. 1983). This study, however, was underpowered to
detect benefits of the size observed.
The sample sizes of the studies that assessed safety were too small to detect less common
side effects. While it appears safe (Level IV evidence), there is little evidence to support
the effectiveness of tranexamic acid. Therefore, the literature does not provide enough
information on which to change the consensus-based recommendations.
Table 12.
Effectiveness of tranexamic acid
Study
Level and quality
of evidence
(Schiavoni et al.
1983)
Level II:
randomised
controlled trial
Quality score:
13/27
External validity
low
Bias minimised
Confounding
avoided
Reporting poor
Population
26 patients, 17
severe
haemophilia A, 2
mild haemophilia
A, 7 severe
haemophilia B
Effectiveness of tranexamic acid
Tranexamic acid 25 mg/kg
3x daily as prophylaxis
Placebo
11 traumatic bleeds
14 traumatic bleeds
6 spontaneous bleeds
4 spontaneous bleeds
589 units FVIII used per
bleeding episode
825 units FVIII used per
bleeding episode
533 units FIX used per
bleeding episode
637 units FIX used per
bleeding episode
No statistically significant differences between groups
reflecting small sample sizes
FVIII= factor VIII, FIX= factor IX
48
Use of recombinant and plasma-derived Factor VIII and IX
Table 13.
Safety of tranexamic acid
Study
Level and quality of
evidence
(Sindet-Pedersen et
al. 1988)
Level IV: case series
Quality score: 1.5/3
Measurement bias
partially minimised
Selection bias
possible
Population
Adverse events to tranexamic acid
15 patients, 13
haemophilia A, 2
haemophilia B
Tranexamic acid orally (25 mg/kg 4x daily) or
as mouthwash (10 ml of 5% tranexamic acid
solution, 4x daily for 2 minutes)
21 occasions of gingival
haemorrhage
No adverse events reported
4 patients with
menorrhagia, 1 Type 1
vWD, 2 Type 2A vWD, 1
Type 2B vWD, aged 17–
42 years. Previous
treatments with DDAVP,
oral contraceptives and
regular dosing with
tranexamic acid had
failed
Single dose tranexamic acid (4 g for 3-5 days)
2 patients with
menorrhagia, 1 Type 1
vWD, 2 Type 2A vWD,
aged 23–43 years
Single dose tranexamic acid (3 g for 1-5 days)
Follow-up adequate
Uncontrolled
(Ong et al. 1998)
Level IV: case series
Quality score: 1/3
Measurement bias
possible
Selection bias
possible
Follow-up adequate
Uncontrolled
(Mohri 2002)
Level IV: case series
Quality score: 1/3
Measurement bias
possible
1 patient complained of occasional diarrhoea
No adverse events reported
Selection bias
possible
Follow-up adequate
Uncontrolled
DDAVP=1-deamino-D-arginine vasopressin; vWD= von Willebrand Disease
Evidence-based clinical practice guidelines
There was no evidence on which to base changes to the previous consensus-based recommendations.
Dosing regimens provided in the current guidelines are consistent with manufacturer’s
recommendations. No paediatric dosages are provided.
Use of recombinant and plasma-derived Factor VIII and IX
49
Fibrin glue
There were no available studies that satisfied the inclusion criteria to assess the
effectiveness of fibrin glue, although two studies provided data on safety. While
conclusions need to be interpreted with caution as the sample sizes in the included
studies were too small to report less common side effects, fibrin glue appears to be a very
safe product, without any side effects, that can be used in conjunction with other
products (Level IV evidence).
Table 14.
Safety of fibrin glue
Study
Level and quality of
evidence
Population
Adverse events after use of fibrin glue
(Avanoglu et al.
1999)
Level IV: prospective
case series
22 patients, 21
haemophilia A, 1
haemophilia B, aged
6–17 years
Fibrin glue used with pdFVIII or pdFIX and
tranexamic mouth wash
Quality score: 2/3
Measurement bias
unlikely
Selection bias
possible
22 circumcisions, 11
with fibrin glue, 11
without
One bleeding episode due to erection
No other adverse events noted
Follow up uncertain
Uncontrolled
(Rakocz et al. 1993)
Level IV: case series
Quality score: 2/3
Measurement bias
unlikely
Selection bias
possible
Follow-up adequate
Uncontrolled
80 patients with
bleeding disorders,
37 severe
haemophilia A/B, 4
mild haemophilia
A/B, 10
mild/moderate vWD,
29 other
135 dental
extractions
Fibrin glue used with aprotinin concentration
1000 KIU/ml
Secondary bleeding in 9/12 severe haemophilia
patients due to insufficient effectiveness
Fibrin glue used with aprotinin concentration
10,000 KIU/ml and tranexamic acid mouthwash
No adverse events
KIU= Kallikrein Inhibitor Units; vWD= von Willebrand Disease
Prothrombin Complex Concentrates (PCCs)
Prior to the development of high-purity plasma derived factor IX and recombinant
factor IX, PCCs were used predominantly for the treatment of haemophilia B patients.
Given the availability of safer products, PCCs are now used rarely in patients without
inhibitors, as they have been associated with an increased incidence of thrombotic events,
such as disseminated intravascular coagulation (DIC) and thromboembolism (Scharrer
1995). Moreover, they are made from pooled human plasma, which has a theoretical risk
of viral transmission. For evaluation of studies on the use of PCCs in patients with
inhibitors, see section on Inhibitors and Tolerisation.
50
Use of recombinant and plasma-derived Factor VIII and IX
Treatment of Bleeding Episodes
Frequent, persistent and prolonged haemorrhage, either spontaneous or trauma-induced,
is the main indication requiring immediate attention in patients with coagulation
disorders. Treatment may take place in a variety of settings—emergency rooms, clinics,
specialist haemophilia care centres and home treatment. The development and
introduction of recombinant factors obviates the need to rely on plasma as the sole
source of antihaemophilic factors and provides a potentially unlimited resource.
For an evaluation of the treatment of bleeding episodes in haemophilia A or B patients
with inhibitors, see section on Inhibitors and Tolerisation.
Haemophilia A, without inhibitors
In Australia, the factor replacement products used for the treatment of patients with
haemophilia A, without inhibitors, are recombinant or plasma-derived factor VIII
products. The recombinant factor VIII (rFVIII) products are: Recombinate™ (Baxter),
Helixate® and Kogenate® (Bayer), and ReFacto® (Wyeth Pharmaceuticals). The only
plasma-derived factor VIII product available in Australia is Biostate® (CSL). These
factors, which vary due to differences in manufacturing processes, are detailed above (see
section on Selection of Products).
Previous Australian consensus-based guidelines
•
Haemophilia A (without inhibitors): Recombinant factor VIII is the treatment of choice for all
patients.
•
Mild-moderate haemophilia A (without inhibitors): Intravenous DDAVP (0.3 mcg/kg) may be
considered. Use with caution in elderly patients and pregnant women.
See Appendix G for details (Haemophilia Foundation Australia Medical Advisory Panel 2000)
Are treatments effective?
Thirteen studies—ranging from Level II to Level IV evidence—investigated the
effectiveness of rFVIII concentrates in haemophilia A patients.
In general, the effectiveness of treatments for haemophilia (A or B) is measured using
specific pharmacokinetic analyses, including the factor coagulant activity, half-life and
response or recovery. These indicate the rate at which the plasma factor levels rise after
treatment. The commonly used measures of factor activity are defined as follows:
Use of recombinant and plasma-derived Factor VIII and IX
51
FVIII:C*
Coagulant activity of FVIII—refers to specific levels or concentration of plasma FVIII.
eg FVIII:C<1 IU/dL = severe haemophilia A
IU = International Units—1 IU, as defined by the World Health Organization standard for human blood
coagulation FVIII is approximately equal to the level of FVIII activity found in 1 mL of fresh pooled
human plasma (Bayer Healthcare). 1 IU FVIII/kg body weight is expected to raise plasma FVIII
activity by 2%. A ratio of actual to predicted FVIII recovery ≥0.66 is considered within normal limits
(Bray et al. 1994).
Incremental Recovery
Increase of FVIII:C/Actual dose (IU) of FVIII/body weight (kg) (Yoshioka et al. 2001)
In vivo recovery (%)
(FVIII:C after infusion of FVIII – FVIII:C before infusion of FVIII (IU/dL)/ expected rise in plasma
FVIII:C x 100 (Yoshioka et al. 2001)
*Official abbreviation for the factor VIII functional assay is now FVIII (Kasper 2004)
To obtain plasma FVIII recovery data, blood samples are drawn before infusion of
factor concentrate (to obtain a baseline measure) and 10 minutes after infusion for
comparison. Clearly, this procedure is less likely to occur for more serious acute bleeding
episodes and/or in young children. Therefore, the available studies provide data on
FVIII recovery from patients in a non-bleeding state and include a mixture of patients
undergoing treatment for bleeding episodes, prophylaxis, surgery or dental procedures.
Comparison of recombinant products
One good quality randomised double-blinded crossover trial (Level II evidence) showed
equivalent efficacy between a newly developed recombinant antihaemophilic factor
(rAHF-PFM) and a recombinant FVIII (Recombinate™). The former was produced by a
plasma/albumin-free method and the latter was processed by a better known method
using genetically engineered cells cultured in the presence of human/animal plasma
proteins or stabilised with human albumin (Tarantino et al. 2004).
Comparison of recombinant and plasma-derived products
One good quality randomised crossover trial (Level II evidence) showed the mean FVIII
activity was equivalent after infusion of either plasma-derived or recombinant factor
concentrate (Fijnvandraat et al. 1997; Berntorp 1997). In three studies, including one
study conducted in a paediatric population (Kelly et al. 1997), there appeared to be an
advantage in FVIII recovery after infusion with rFVIII compared to pdFVIII in nonbleeding previously treated patients (PTPs) (Kelly et al. 1997; Morfini et al. 1992;
Schwartz et al. 1990) (Level III-1 and III-2 evidence) (Table 15). However, inadequate
randomisation or incomplete blinding of outcomes in the lower level studies may have
yielded an overestimate of effect.
Overall, the higher-level evidence indicates that there is no difference or, at the most, a
slightly better factor VIII recovery after infusion with rFVIII compared to pdFVIII
(Table 15).
52
Use of recombinant and plasma-derived Factor VIII and IX
Table 15.
FVIII recovery following treatment with rFVIII in haemophilia A patients
Study
(Tarantino et al.
2004)
Level and quality of
evidence
Population
Level II: double-blinded
randomised crossover
trial
56 PTPs with
moderate-severe
haemophilia A, aged
10–65 years, without
inhibitors, with ≥150
exposure days
rAHF-PFMa (50
IU/kg)
rFVIII
(50 IU/kg)
% change
2.6±0.5
2.4±0.5
8.3%
12 PTPs with severe
haemophilia A, aged
17–64 years, without
inhibitors
rFVIII SQb
(50 IU/kg)
pdFVIII
(50 IU/kg,
Octonativ)
% change
2.4±0.21
2.4±0.26
0
10 non-bleeding PTPs
with severe
haemophilia A, aged
6–12, without
inhibitors
rFVIII
(50 IU/kg,
RecombinateTM)
pdFVIII
(50 IU/kg,
Hemofil®
M)
% change
1.91±0.14%
1.5±0.15%
27.3%
Quality score: 25/27
Selection bias
minimised
Incremental FVIII recovery (IU/kg-1)
NS
Confounding avoided
Follow-up adequate
Patients representative
(Fijnvandraat et al.
1997; Berntorp
1997)
Level II: randomised
single-blinded (patient)
crossover trial
Quality score: 21/27
Selection bias
minimised
Confounding avoided
Follow-up adequate
Patients representative
(Kelly et al. 1997)
Level III-1: quasirandomised singleblinded (investigator)
controlled trial
Quality score: 21/27
Clinical importancee:
2/4
p=0.007
Selection bias
minimised
Confounding avoided
Follow-up adequate
Patients representative
(White et al. 1997)
Level III-2: nonrandomised crossover
trial
Quality score: 20/27
69 PTPs with
moderate-severe
haemophilia A,
without inhibitors
Selection bias unclear
rFVIII
(50 IU/kg,
RecombinateTM)
pdFVIII
(50 IU/kg,
Hemofil®
M)
% change
2.40±0.97
2.47±0.33
2.9%
p=0.59
Confounding avoided
Follow-up adequate
Patients representative
(Schwartz et al.
1990; Seremetis et
al. 1999)
Level III-2: nonrandomised crossover
trial
Quality score: 19/27
Clinical
3/4
17 non-bleeding PTPs
with moderate-severe
haemophilia A,
without inhibitors
importancee:
rFVIII
(50 IU/kg)
pdFVIII
(50 IU/kg,
Koate HS)
% change
2.68±0.52
2.42±0.33
10.7%
p=0.026
Selection bias unclear
Confounding avoided
Follow-up adequate
Patients representative
Use of recombinant and plasma-derived Factor VIII and IX
(cont.)
53
Table 15 (cont.)
Study
(Morfini et al. 1992)
Level and quality of
evidence
Level III-2: nonrandomised crossover
trial
Population
47 PTPs with severe
haemophilia A,
without inhibitors
Quality score: 16/27
Clinical importancee:
3/4
Incremental FVIII recovery (IU/kg-1)
rFVIII
(50 IU/kg,
RecombinateTM)
pdFVIII
(50 IU/kg,
Hemofil® M)
% change
2.31c
1.99c
16.1%
In vivo recovery (%)
107.1±45.6
Selection bias possible
92.2±32.0
14.9%
p<0.019
Confounding avoided
Follow-up adequate
Patients representative
(Arkin et al. 1991)
Level III-3: historical
control study
Quality score: 5/27
Selection bias likely
Confounding likely
Follow-up uncertain
14 non-bleeding PTPs
with moderate-severe
haemophilia A, aged
4–72, without
inhibitors
rFVIII
(20–40 IU/kg, 3 x
week)
pdFVIII
% change
2.7±0.6
2.5±0.33
8.0%
p=0.29
Historical controls: no
description provided
External validity
uncertaind
(Aygoren-Pursun et
al. 1997)
Level IV: uncontrolled
before-and-after study
Quality score: 2/3
Selection bias uncertain
Follow-up adequate
39 PTPs with mildsevere haemophilia A,
aged 2–62 years,
treated for bleeding
episodes, prophylaxis
and surgery
rFVIII (50 IU/kg,
Kogenate®)
Month 0
2.4±0.83
Month 12
2.12±0.61
Uncontrolled
(Yoshioka et al.
2001)
Level IV: case series
Quality score: 2/3
Selection bias uncertain
Follow-up adequate
Uncontrolled
20 PTPs with
moderate-severe
haemophilia A, aged
12–55 years, without
inhibitors, treated for
bleeding episodes
rFVIII-FSd (10-50 IU/kg,
Kogenate®)
Week 0
1.73±0.40
Week 12
1.84±0.40
Week 24
1.95±0.55
recombinant antihaemophilic factor, plasma albumin-free; recombinant B-domain deleted FVIII; calculated by authors from data provided; d
recombinant FVII formulated with sucrose; e Rank scores for the clinical importance of the benefit/harm (1/4 ranked highest and 4/4 ranked
lowest); d Information on population source not provided; MTPs = minimally treated patients; PTPs = previously treated patients; PUPs =
previously untreated patients
a
b
c
Effectiveness of treatment of bleeding episodes with rFVIII was also assessed according
to the number of infusions needed to control haemostasis. Seven before-and-after
studies or case series (Level IV evidence) consistently reported that approximately 90%
of bleeding episodes were managed with 1–2 infusions and were rated as excellent or
good by patients or physicians, using a four- or five-point Likert scale, ranging from
excellent to no or less effect (Table 16).
54
Use of recombinant and plasma-derived Factor VIII and IX
Table 16.
Number of infusions required to control haemostasis in haemophilia A patients
Study
(Gringeri et al. 2004)
Level and quality of
evidence
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Population
Control of haemostasis
Number of infusions
per bleeding
episode
% of bleeding
episodes
25 PTPs with severe
haemophilia A, aged
6–60 years, without
inhibitors, treated for
bleeding episodes
1
2
3
4
>4
76.7
12.2
6.6
2.8
1.7
58 PTPs with
moderate–severe
haemophilia A, without
inhibitors, treated
1
2
≥2
82.0
12.0
6.0
39 PTPs with mildsevere haemophilia A,
aged 2–62 years,
treated with rFVIII
(Kogenate®) for
bleeding episodes,
prophylaxis and
surgery
1
2
3
4
≥5
75.5
14.5
5.6
1.4
3.1
43 PUPs and MTPs
with mild-severe
haemophilia A, aged
3–386 months, treated
with rFVIII
(Kogenate®) for
bleeding episodes
1
2
3
98.5
1.4
0.1
102 PUPs with mildsevere haemophilia A,
without inhibitors,
treated for bleeding
episodes, prophylaxis
or surgery
1
2
3
4
≥5
82.2
10.9
3.2
1.3
2.3
20 PTPs with
moderate-severe
haemophilia A, aged
12–55 years, without
inhibitors, treated for
bleeding episodes
Mean number of
infusions per bleeding
episode
1.4±1.2
1-2
88.1
Follow-up adequate
Uncontrolled
(Schwartz et al. 1990;
Seremetis et al. 1999)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Aygoren-Pursun et al.
1997)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Yoshioka et al. 2003)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Lusher et al. 2004)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Yoshioka et al. 2001)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Giangrande 2002)
Level IV: case series
Quality score: 1/3
Selection bias
uncertain
Follow-up uncertain
Uncontrolled
31 PUPs and MTPs
with severe
haemophilia A, aged ≤
4 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
MTPs = minimally treated patients; PTPs = previously treated patients; PUPs = previously untreated patients
Use of recombinant and plasma-derived Factor VIII and IX
55
Are treatments safe?
In the early 1980s, up to 80% of haemophilia patients became infected with HIV through
transfusion of infected plasma-derived products. Since then, the safety of plasma-derived
factor concentrates has improved substantially with the introduction of two virus
inactivation procedures: 1) solvent/detergent treatment, which inactivates lipidenveloped viruses; and 2) heat treatment, which targets non-lipid enveloped viruses
(Ananyeva et al. 2004). There have been no reports of HIV in Australia since the
introduction of heat and solvent/detergent (SD) treatment of coagulation factor
concentrates (Haemophilia Foundation Australia Medical Advisory Panel 2000).
More recently, nanofiltration, which filters out lipid-enveloped viruses and prions, has
been developed. For example, the Pall Corporation has developed membrane filtration
technology to remove prions, such as those associated with variant Creuztfeldt-Jakob
disease (vCJD) in humans and Bovine Spongiform Encephalopathy in cattle (Pall
Corporation 2004). Late stage testing of the Leukotrap® Affinity prion reduction filter
has shown effective removal of different strains of infectious prions, including vCJD and
scrapie, from red blood cell concentrates. However, there remains a theoretical risk that
previously unrecognised infective agents may still gain access to the donor pool. For
example, although there has been no reported transmission of blood-borne viral agents
in Australia since 1990 (JBC FVIII/FIX working party & National Blood Authority
2004), confidence in plasma-derived products was seriously undermined in Japan when
1) plasma-derived products produced from pooled plasma were withdrawn after donors
were diagnosed with Creutzfeld Jacob disease; and 2) evidence of hepatitis A
transmission through pdFVIII products was revealed (Yoshioka et al. 2001). Therefore,
methods used to purify plasma-derived products are a direct function of the accuracy of
information concerning novel infectious agents.
Recombinant products, which contain no components from the donor pool, are believed
to offer substantial protection from viral transmission. However, the use of human
albumin as a stabiliser during the production of recombinant products has the theoretical
potential to introduce pathogens, such as prions or unknown agents, into these products.
Only the complete exclusion of all human or animal proteins would eliminate that
possibility. Recombinant products are continuously in development and the potential
immunogenicity of recombinant factors is still in question. Further, the transfer of
patients to different blood coagulation products was thought to increase the potential
risk for inhibitor development following a cluster of cases with inhibitors after patients
were switched to products that had undergone newly developed viral inactivation
processes (Ewenstein et al. 2004). The viral inactivation methods were thought to have
modified the tertiary structure of the FVIII molecule, thereby exposing neoantigens.
The key safety outcomes reported in the studies included in this review are: the
transmission of viral infections, the development of inhibitors to FVIII and adverse
reactions to the treatment. Several studies also reported immunogenic responses to
proteins used in the recombinant manufacturing process, including murine
immunoglobulins and hamster proteins, and seroconversions for human
immunodeficiency virus (HIV), hepatitis (A, B, or C) or parvovirus (B19).
Transmission of viral infections
One good quality retrospective cohort study (Level III-2 evidence) evaluated the risk of
human parvovirus B19 transmission in rFVIII, a combination of rFVIII and pdFVIII,
56
Use of recombinant and plasma-derived Factor VIII and IX
and pdFVIII alone (Soucie et al. 2004). The patients who received pdFVIII had 7.6 times
the odds of B19 antibody seropositivity compared to patients who had never received
blood or blood components (OR= 7.6, 95% CI 3.6, 15.9). Patients who received only
rFVIII had an equivalent risk to patients who had never received blood or blood
components (OR= 0.8, 95% CI 0.4, 1.5) (Level III-2 evidence).
No included studies reported new cases of viral transmission such of HIV, hepatitis A, B
or C, after treatment with either plasma-derived or recombinant factor concentrate. In
addition, there were no reports of adverse reactions in patients with a positive antibody
reaction to animal proteins, even in cases with detectable pre-existing antibodies to these
proteins (White et al. 1997) (Level IV evidence).
One average quality historically controlled study (Level III-3 evidence) examined the
immune status of HIV seropositive and seronegative haemophilia A patients treated with
rFVIII (Mannucci et al. 1994). The percentage of CD4 and CD8 cells, a surrogate
measure of HIV infection, showed no significant change in seronegative or seropositive
patients over a period of 3.5 years. However, a small but significant decrease in the
absolute count of CD4 cells was apparent in HIV seropositive patients (p=0.001). The
clinical significance of this difference is unclear.
Development of inhibitors
Inhibitor detection is conducted using the Bethesda assay, with or without the Nijmegen
modification (Verbruggen et al. 1995), and results are expressed in Bethesda units (BU)g.
If the inhibitor titre is high (>5 BU/ml), factor replacement therapy is ineffective and
bleeding persists. With low titre inhibitor (<5 BU/ml), haemostasis may be achieved with
higher doses. Patients with severe haemophilia A with high-titre inhibitors are most at
risk for recurrent bleeds and chronic haemarthroses.
The best available evidence to determine the rate of inhibitor development in
recombinant compared to plasma-derived products was provided by one average quality
non-randomised controlled trial (Level III-2 evidence) in which patients were transferred
from plasma-derived factors to recombinant factor products (Giles et al. 1998) and two
good-average quality case series (Level IV evidence) in patients who had previously been
treated with plasma-derived factors (Gringeri et al. 2004). After two years and six months
follow-up, respectively, neither study showed evidence of increased incidence in FVIII
inhibitors following transfer. Results are shown in Table 17.
Of four case series that included patients previously treated with pdFVIII, two studies
reported low-level transient inhibitors in one patient (0.9% and 1.4%) (Schwartz et al.
1990; Tarantino et al. 2004; White et al. 1997; Yoshioka et al. 2001). One patient had
severe haemophilia (Tarantino et al. 2004) while the other had a history of inhibitor
development with use of pdFVIII (White et al. 1997). However, previously treated
patients (PTPs) without inhibitors are thought to be at lower risk of inhibitor
development—compared to previously untreated patients (PUPs)—since PTPs failed to
develop inhibitors with previous replacement therapy (Lusher et al. 1993; Yoshioka et al.
2003).
g Bethesda units (BU) = a measure of inhibitor activity – the amount of inhibitor that inactivates 50% or
0.5 units of a coagulation factor during the incubation period
Use of recombinant and plasma-derived Factor VIII and IX
57
One poor quality historically controlled study (Level III-3 evidence) and six case series
(Level IV evidence) assessed the development of inhibitors in PUPs or minimally treated
patients (MTPs)h following treatment with rFVIII. Slightly higher levels of both high-titre
and low-titre inhibitors were detected in pdFVIII-treated patients compared to rFVIIItreated patients. However, results from this study are subject to considerable selection
and measurement biases.
In the lower level evidence (Level IV evidence) studies that assessed development of
inhibitors in haemophilia A patients receiving rFVIII products, the proportion of
patients developing high or low titre inhibitors varied widely across studies (0–23%). The
characteristics of patients (severity of disease, previous treatments), factor products, and
differences in study methodology (length of follow-up, frequency of inhibitor testing)
may influence the reported rate of inhibitor development. For example, transient or low
concentration inhibitors were more likely to be detected if patients were tested for
inhibitors—whether clinically indicated or not—compared to studies that performed
tests only when presence of inhibitors was clinically indicated. The latter studies tend to
underestimate the incidence of inhibitor formation (Lusher et al. 1993).
Therefore, since the available evidence indicates that recombinant factors, which provide
a higher safety profile with respect to viral transmission, do not induce higher rates of
inhibitor development, they should be the first line of treatment (Level III-2 evidence).
MTPs—minimally treated patients who have received ≤ 2 prior infusions with either recombinant or
plasma-derived products
h
58
Use of recombinant and plasma-derived Factor VIII and IX
Table 17.
Development of inhibitors in haemophilia A patients treated with recombinant factor VIII
Study
Level and quality of
evidence
Population
No. of patients with inhibitor development
872 PTPs with mild–
severe haemophilia,
transferred from
plasma-derived
products to
recombinant or affinitypurified FVIII
rFVIII
(N=339)
AP-FVIII
(N=32)
High-titre
inhibitors
17 (5.0%)
2 (6.3%)
Low-titre
inhibitors
52 (15.3%)
5 (15.6%)
Patients transferred from pdFVIII to rFVIII
(Giles et al. 1998)
Level III-2: non
randomised controlled
trial
Quality score: 15/27
Selection bias not
minimised (patient
self-selection)
Confounding unclear
Follow-up adequate
External validity
uncertaina
(Gringeri et al. 2004)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
25 PTPs with severe
haemophilia A, aged
6–60 years, without
inhibitors, treated for
bleeding episodes
High-titre inhibitor
Low-titre inhibitor
1 (4.0%)
0
94 PTPs with mild–
severe haemophilia A,
without inhibitors,
treated for bleeding
episodes or
prophylaxis
High-titre inhibitor
Low-titre inhibitor
1 (1.1%)
0
Follow-up adequate
Uncontrolled
Level IV: case series
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
59
Table 17 (cont.)
Previously treated patients (PTPs)
(Tarantino et al. 2004)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Confounding avoided
108 PTPs with
moderate-severe
haemophilia A, aged
10–65, without
inhibitors, with ≥150
exposure days
High-titre inhibitor
Low-titre inhibitor
0
1 (0.9%) transient
69 PTPs with
moderate–severe
haemophilia A, without
inhibitors
High-titre inhibitor
Low-titre inhibitor
0
1 (1.4%) transient
58 PTPs with
moderate–severe
haemophilia A, without
inhibitors, treated
None detected
20 PTPs with
moderate–severe
haemophilia A, aged
12–55 years, without
inhibitors, treated for
bleeding episodes
None detected
Follow-up adequate
Uncontrolled
(White et al. 1997)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Confounding avoided
Follow-up adequate
Uncontrolled
(Schwartz et al. 1990;
Seremetis et al. 1999)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Yoshioka et al. 2001)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
Previously untreated patients (PUPs)
(Kreuz et al. 2002)
Level III-3: historical
control study
Quality score: 11/27
Selection bias likely
Confounding possible
72 PUPs with
moderate–severe
haemophilia A, without
inhibitors, treated for
bleeding episodes,
prophylaxis, or surgery
Follow-up unclear
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
(Lusher et al. 1993)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
60
pdFVIII
(N=51)
High-titre
inhibitors
4 (19.0%)
13 (25.5%)
Low-titre
inhibitors
0
5 (9.8%)
4 (19.0%)
18 (35.3%)
Total
Patients
representative
(Lusher et al. 2004)
rFVIII
(N=21)
102 PUPs with mild–
severe haemophilia A,
without inhibitors,
treated for bleeding
episodes, prophylaxis
or surgery
High-titre inhibitor
Low-titre inhibitor
12 (11.8%)
9 (8.8%)
8 transient
95 PUPs with mild–
severe haemophilia A,
aged 0–658 months,
treated for bleeding
episodes or
prophylaxis
High-titre inhibitor
Low-titre inhibitor
5 (5.3%)
11 (11.6%)
4 transient
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 17 (cont.)
Study
(Bray et al. 1994)
Level and quality of
evidence
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
(Rothschild et al.
1998)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Population
Inhibitor development associated with factor
replacement therapy
73 PUPs with
moderate–severe
haemophilia A, aged 2
days–50 months,
without inhibitors,
treated for bleeding
episodes or
prophylaxis
High-titre inhibitor
Low-titre inhibitor
8 (11.0%)
9 (12.3%)
5 transient
50 PUPs with severe
haemophilia A, aged
1–43 months, treated
for bleeding episodes
or prophylaxis
High-titre inhibitor
Low-titre inhibitor
7 (14.0%)
7 (14.0%)
2 transient
43 PUPs and MTPs
with mild–severe
haemophilia A, aged
3–386 months, treated
with rFVIII
(Kogenate®) for
bleeding episodes
High-titre inhibitor
5 (11.6%)
all severe
10 (23.3%)
8 severe
2 moderate
7 transient
31 PUPs and MTPs
with severe
haemophilia A, aged ≤
4 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
High-titre inhibitor
Low-titre inhibitor
Follow-up adequate
(Yoshioka et al. 2003)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
(Giangrande 2002)
Level IV: case series
Quality score: 1/3
Selection bias
uncertain
Follow-up uncertain
Low-titre inhibitor
1 (3.2%)
3 (9.7%)
2 transient
a Information on population source not provided; AP-FVIII= affinity-purified factor VIII; MTPs = minimally treated patients; PTPs = previously
treated patients; PUPs = previously untreated patients
Eight low level studies (Level IV evidence) measured the number of days of exposure
(ED) to factor concentrates prior to inhibitor development. The median ED varied
between five and 17 days across studies (Table 18). It is suggested that inhibitors be
tested for no earlier than three days after administration of factor concentrates, or when
the expected response is absent (Level IV evidence).
Use of recombinant and plasma-derived Factor VIII and IX
61
Table 18.
Exposure days prior to inhibitor development in haemophilia A patients treated with
recombinant factor VIII
Study
Level and quality of
evidence
Population
Number of exposure days (ED) prior to
inhibitor development
Inhibitor positive
Median ED (range)
25 PTPs with severe
haemophilia A, aged
6–60 years, without
inhibitors, treated for
bleeding episodes
1 (4.0%)
5
94 PTPs with mild–
severe haemophilia A,
without inhibitors,
treated for bleeding
episodes or
prophylaxis
1 (1.1%)
10
102 PUPs with mild–
severe haemophilia A,
without inhibitors,
treated for bleeding
episodes, prophylaxis
or surgery
21 (20.6%)
9 (3–54)
73 PUPs with
moderate–severe
haemophilia A, aged 2
days–50 months,
without inhibitors,
treated for bleeding
episodes or
prophylaxis
17 (23.9%)
9 (3–45)
50 PUPs with severe
haemophilia A, aged
1–43 months, treated
for bleeding episodes
or prophylaxis
14 (28.0%)
17 (3–69)
43 PUPs and MTPs
with mild–severe
haemophilia A, aged
3–386 months, treated
with rFVIII (Kogenate)
for bleeding episodes
15 (34.9%)
12 (1–48)
72 PUPs with
moderate–severe
haemophilia A, without
inhibitors, treated for
bleeding episodes,
prophylaxis, or surgery
22 (31%)
15 (4–195)
Previously treated patients (PTPs)
(Gringeri et al. 2004)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
Level IV: case series
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
Previously untreated patients (PUPs)
(Lusher et al. 2004)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Bray et al. 1994)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Rothschild et al.
1998)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Yoshioka et al. 2003)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Kreuz et al. 2002)
Level IV: case series
Quality score: 1/3
Selection bias likely
Confounding possible
Follow-up unclear
Uncontrolled
62
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 18 (cont.)
Study
Level and quality of
evidence
(Giangrande 2002)
Level IV: case series
Quality score: 1/3
Selection bias
uncertain
Follow-up uncertain
Uncontrolled
Population
31 PUPs and MTPs
with severe
haemophilia A, aged ≤
4 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
Number of exposure days (ED) prior to
inhibitor development
Inhibitor positive
Median ED (range)
4 (12.9%)
8 (3–12)
MTPs = minimally treated patients; PTPs = previously treated patients; PUPs = previously untreated patients
Adverse events and complications
A broad range of adverse events were found to be associated with rFVIII infusions—but
in a relatively small proportion of patients (<1.0%). Reactions were mild to moderate in
severity, transient or reversible, and none resulted in discontinuation of treatment.
Results are shown in Table 19.
Table 19.
Adverse events in haemophilia A patients treated with recombinant factor VIII
Study
(Tarantino et al. 2004)
Level and quality of
evidence
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Confounding avoided
Population
Adverse events
Ratio of adverse
events to total
number of
infusions
108 PTPs with
moderate–severe
haemophilia A, aged
10–65, without
inhibitors, with ≥150
exposure days
19 mild–moderate
adverse events related to
AHF-PFM:
Adv. events = 19
Taste perversion,
headache, fever,
diarrhoea, dizziness, hot
flushes, pain (upper
abdomen, lower chest),
shortness of breath,
sweating, nausea, itching
Ratio = 0.15%
58 PTPs with
moderate–severe
haemophilia A, without
inhibitors, treated for
bleeding episodes or
surgical procedures
42 mild-moderate adverse
events associated with
Kogenate®:
Adv. events =42
Burning/erythema at
infusion site, nausea,
pruritis, dizziness, rash,
dyspnea, chest tightness,
dry mouth
Ratio = 0.23%
102 PUPs with mild–
severe haemophilia A,
without inhibitors,
treated for bleeding
episodes, prophylaxis
or surgery
16 mild–moderate
adverse events
associated with
Kogenate®:
Adv. events =16
Follow-up adequate
Patients
representative
Uncontrolled
(Schwartz et al. 1990;
Seremetis et al. 1999)
Level IV: case series
Quality score: 3/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Lusher et al. 2004)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
Use of recombinant and plasma-derived Factor VIII and IX
Urticaria, flushing,
erythema, rash
Total number of
infusions = 12,597
Total number of
infusions =17,922
Total number of
infusions =13,464
Ratio = 0.12%
(cont.)
63
Table 19 (cont.)
Level IV: case series
(Lusher et al. 1993)
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
95 PUPs with mild–
severe haemophilia A,
aged 0–658 months,
treated for bleeding
episodes or
prophylaxis
3 minor adverse events
associated with
Kogenate®:
Adv. Events =3
Urticaria, flushing,
erythema at infusion site
Ratio = 0.09%
73 PUPs with
moderate–severe
haemophilia A, aged 2
days–50 months,
without inhibitors,
treated for bleeding
episodes or
prophylaxis
1 transient adverse event,
which existed prior to
infusion, worsened by
RecombinateTM:
Adv events =1
69 PTPs with
moderate–severe
haemophilia A, without
inhibitors
13 mild transient adverse
events:
39 PTPs with mild–
severe haemophilia A,
aged 2–62 years,
treated for bleeding
episodes, prophylaxis
and surgery
3 mild and transient
adverse events
associated with
Kogenate®:
Total number of
infusions =3,315
Uncontrolled
(Bray et al. 1994)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(White et al. 1997)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Confounding avoided
Erythematous rash
Slight flushing, nausea,
tiredness, epistaxis, brief
dizziness
Total number of
infusions =1,785
Ratio = 0.06%
Adv events =13
Total number of
infusions =13,591
Ratio = 0.10%
Follow-up adequate
Uncontrolled
(Aygoren-Pursun et al.
1997)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Flushing, paraesthesia at
infusion site, headache
Adv. Events =3
Total number of
infusions =3,679
Ratio = 0.08%
Uncontrolled
AHF-PFM = antihaemophilic factor, plasma/albumin free; PTPs = previously treated patients; PUPs = previously untreated patients
Overall, the data evaluated in this review suggest that rFVIII is safe and effective for the
treatment of haemophilia A patients (without inhibitors) and that inhibitor development
is equally likely with rFVIII treatment or prophylaxis as with pdFVIII.
Evidence-based clinical practice guidelines
Based on the evidence, it is recommended that bleeding episodes be treated according to the existing
guidelines, with modifications:
•
Haemophilia A: Recombinant FVIII is the treatment of choice for patients with haemophilia A
without inhibitors to FVIII (Level II evidence for effectiveness; Level III-2 evidence for safety).
Inhibitor development is equally likely with rFVIII treatment or prophylaxis, as with pdFVIII (Level
III-2 evidence).
•
Patients should be tested for inhibitors no earlier than day three after initial administration of factor
concentrate, particularly in young children and/or those with severe haemophilia A. Given the wide
range in exposure days prior to inhibitor development, re-testing should be performed at regular
intervals (Level IV evidence) or when expected response is absent (expert opinion).
64
Use of recombinant and plasma-derived Factor VIII and IX
Haemophilia B, without inhibitors
In Australia, the product used for treatment of patients with haemophilia B, without
inhibitors, is recombinant factor IX (rFIX), BeneFIX® (Wyeth Pharmaceuticals). A
purified plasma-derived FIX concentrate, MonoFIX-VF (CSL) is also available.
Previous Australian consensus-based guidelines
Haemophilia B: High purity FIX concentrates are preferred over prothrombin complex concentrates
(PCCs).
See Appendix G for details (Haemophilia Foundation Australia Medical Advisory Panel 2000)
Are treatments effective?
Several studies (Bjorkman et al. 2001; Roth et al 2001a; White et al. 1998) provided data
on the same group of patients and, where possible, the most comprehensive and up-todate data were extracted.
Two good-to-average quality randomised crossover trials (Level II evidence) (White et al.
1998; Ewenstein et al. 2002) and one average quality historically controlled study (Level
III-3 evidence) (Poon et al. 2002) showed lower factor recovery after rFIX infusion than
for pdFIX in patients with haemophilia B (p<0.0001) (Table 20). The reduction in FIX
recovery with rFIX was not only statistically significant, but also indicated clinical harmi.
Importantly, two studies reported large inter-patient variability in pharmacokinetic
analyses in both rFIX and pdFIX concentrates (Ewenstein et al. 2002; Poon et al. 2002).
In addition, both rFIX and pdFIX showed lower recovery in young children (<16 years)
compared with older children and adults.
However, dosage multiplication factors of 1.57 [95% CI 1.48-1.66] and 1.19 [95% CI
1.13-1.26] were recommended when treating haemophilia B patients aged ≤15 and >15
years, respectively, with rFIX (Poon et al. 2002). Findings from these studies suggest that
patients with haemophilia B should be closely monitored for FIX activity/recovery after
treatment with rFIX concentrate to optimise individual treatment regimens.
Clinical importance of statistically significant effects was ranked according to the checklist provided in
Appendix H. More than 20% reduction in FIX recovery—vs control group FIX recovery—indicated a
clinical harm.
i
Use of recombinant and plasma-derived Factor VIII and IX
65
Table 20.
FIX recovery following treatment of bleeding episodes in haemophilia B patients
Study
Level and quality of
evidence
(Ewenstein et al.
2002)
Level II: randomised
crossover trial
Quality score: 22/27
Clinical
1/4
importancea:
Population
38 non-bleeding PTPs
with severe
haemophilia B, aged
7–75 years, without
inhibitors
Incremental FIX recovery, % IU/kg-1
rFIX
(50 IU/kg)
pdFIX
(50 IU/kg)
% change
p-value
0.86±0.31
1.71±0.73
49.7%
p≤0.0001
rFIX
(50 IU/kg)
pdFIX
(50 IU/kg)
% change
p-value
0.8±0.3
1.2±0.3
33.3%
rFIX (N=126)
(50 IU/kg)
pdFIX (N=74)
(50 IU/kg)
% change
p-value
0.77±0.19
1.05±0.26
26.7%
p<0.0001
Selection bias
minimised
Confounding avoided
Follow-up adequate
Patients
representative
(White et al. 1998)
Level II: randomised
crossover trial
11 PTPs with
haemophilia B
Quality score: 17/27
Clinical importancea:
2/4
Selection bias
minimised
Confounding avoided
Follow-up adequate
External validity
uncertainb
(Poon et al. 2002)
Level III-3: historical
control study
Quality score: 15/27
Clinical
1/4
PTPs with mild–
severe haemophilia B,
aged 1–74 years
importancea:
Selection bias
uncertain
Confounding possible
Follow-up adequate
Patients
representative
(Roth et al. 2001a;
Bjorkman et al. 2001)
Level IV: uncontrolled
before-and-after study
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
56 PTPs with
moderate–severe
haemophilia B, aged
4–56 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
rFIX (50 IU/kg)
Mean FIX activity
increase
0.75 (range 0.34-1.38)
Uncontrolled
Rank scores for the clinical importance of the benefit/harm (1/4 ranked highest and 4/4 ranked lowest); b Information on population source not
provided; PTPs = previously treated patients
a
66
Use of recombinant and plasma-derived Factor VIII and IX
One uncontrolled before and after study (Level IV evidence) showed that over 90% of
bleeding episodes were controlled by 1–2 infusions of rFIX (Roth et al. 2001a; White et
al. 1998) (Table 21).
Table 21.
Number of FIX infusions required to control haemostasis in haemophilia B patients
Study
(Roth et al. 2001a)
(White et al. 1998)
Level and quality of
evidence
Population
Level IV: case series
56 PTPs with
moderate–severe
haemophilia B, aged
4–56 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
Control of haemostasis
Number of infusions
per bleeding episode
% of bleeding
episodes
1
2
3
4
>4
80.9
11.6
3.6
1.8
2.1
PTPs = previously treated patients
Are treatments safe?
Transmission of viral infections
The preparation of rFIX occurs without the use of animal or human plasma-derived
products, thereby eliminating potential transmission of viruses or other known or
unknown pathogens (Roth et al. 2001a). There were no reported cases of transmission of
viral infections in patients treated with rFIX products.
Development of inhibitors
One uncontrolled before-and-after study (Level IV evidence) reported a transient low
titre inhibitor in one patient with severe haemophilia (1.8%) after 39 days of treatment
with rFIX (Roth et al. 2001a) (Table 22 and Table 23).
Table 22.
Development of inhibitors in haemophilia B patients treated with recombinant factor IX
Study
Level and quality of
evidence
Population
(Roth et al. 2001a)
(White et al. 1998)
Level IV: case series
56 PTPs with
moderate–severe
haemophilia B, aged
4–56 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
Inhibitor development associated with
treatment
High-titre inhibitor
Low-titre inhibitor
0
1 (transient)
PTPs = previously treated patients
Use of recombinant and plasma-derived Factor VIII and IX
67
Table 23.
Exposure days prior to inhibitor development in haemophilia B patients treated with
recombinant factor IX
Study
Level and quality of
evidence
Population
(Roth et al. 2001a)
Level IV: uncontrolled
before-and-after study
56 PTPs with
moderate–severe
haemophilia B, aged
4–56 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Number of exposure days (ED) prior to
inhibitor development
Inhibitor positive
Median ED (range)
1 (1.8%)
39
Uncontrolled
PTPs = previously treated patients
Adverse events and complications
The majority of adverse events associated with rFIX were minor allergic reactions (Table
24). Data from the Canadian registry of haemophilia patients reported anaphylactic
reactions to rFIX in two patients after 1–5 years of exposure (Poon et al. 2002).
Table 24.
Adverse events in haemophilia B patients treated with recombinant factor IX
Study
Level and quality of
evidence
Population
(Roth et al. 2001a)
(White et al. 1998)
Level IV: case series
56 PTPs with
moderate–severe
haemophilia B, aged
4–56 years, without
inhibitors, treated for
bleeding episodes,
prophylaxis or surgery
55 adverse events
associated with rFIX:
244 PTPs with mild–
severe haemophilia B
Anaphylactic reaction
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Uncontrolled
(Poon et al. 2002)
Level IV: case series
Quality score: 1/3
Selection bias
uncertain
Confounding possible
Follow-up adequate
Adverse events
Minor allergic reaction
Ratio of adverse
events to total
number of infusions
Adv. Events =55
Total number of
infusions =7362
Ratio = 0.75%
Not estimable
From Canadian
registry data: 2 patients
(of 244) exposed to
rFIX for 1–5 years
developed
anaphylactic reactions
Uncontrolled
PTPs = previously treated patients
Evidence-based clinical practice guidelines
Based on the evidence, it is recommended that bleeding episodes in haemophilia B patients, without
inhibitors, be treated in the following manner:
Haemophilia B: Recombinant FIX is safe for treatment of patients with haemophilia B without inhibitors
(Level IV evidence). Due to the lower rFIX recovery compared to pdFIX, and the large inter-patient
variability, individual dosing regimens should be monitored by FIX recovery/activity assays (Level II
evidence). The recommended rFIX dosage, compared to pdFIX, should be increased by a multiplication
factor of 1.6 for patients aged ≤15 years and 1.2 for patients aged 16 years and over (Level III-3
evidence).
68
Use of recombinant and plasma-derived Factor VIII and IX
Von Willebrand disease
Von Willebrand disease is caused by either quantitative or qualitative abnormalities of
von Willebrand factor (vWF), which is required for coagulation. The three main types of
von Willebrand disease are classified as 1, 2 or 3 and these are further divided into
subtypes. The types listed in Australia on the Australian Bleeding Disorders Registry
(ABDR)j are shown in Table 25.
Table 25.
Type of vWD
1
Number of patients listed on Australian Bleeding Disorders Registry for vWD subtypes
Number of patients on
the ABDR
Description (adapted from Kasper, 2004)
301
vWF is normal in structure; mild-moderate quantitative deficiency of
vWF; ristocetin-induced platelet aggregation is normal
2 (undefined)
24
vWF is abnormal in structure; qualitative deficiency of vWF
2A
10
Decreased platelet-dependent vWF function; absence of high molecular
weight multimers; ristocetin-induced platelet aggregation is reduced or
absent; bleeding time is prolonged; collagen binding is low
2B
11
Increased platelet-dependent vWF function; absence of high molecular
weight multimers; ristocetin-induced platelet aggregation is high at low
concentrations of ristocetin cofactor
2M
16
Decreased platelet dependent vWF function, with normal multimeric
structure; ristocetin cofactor is deficient
2N
3
Decreased vWF affinity for factor VIII
3
21
Patients are unable to synthesise vWF; virtual absence of vWF; usually
severe deficiency; ristocetin cofactor levels are undetectable
Unrecorded
435
Total
821
ABDR= Australian Bleeding Disorders Registry, includes data from all states except NSW; vWD= von Willebrand Disease; vWF= von
Willebrand factor
The goal of therapy in patients with von Willebrand disease is to correct the dual defect
of abnormal platelet adherence and abnormal coagulation due to low factor VIII levels.
The methods of testing the effectiveness of treatments for von Willebrand disease differ
from the methods appropriate with haemophilia. Effectiveness of treatments for von
Willebrand disease may be tested by measuring the vWF antigen, vWF activity—
ristocetin cofactor or collagen binding and bleeding time (See Table 26). High purity
plasma concentrates have been found to return FVIII:Ck to normal, without a reduction
in bleeding time, therefore FVIII:C should not be used as the sole indicator of efficacy
with von Willebrand patients (Berntorp 1994).
Platelet function analysers such as the PFA-100® (Dade) have been used for therapeutic
monitoring of patients with vWD treated with DDAVP or FVIII/vWF concentrates
(Franchini et al. 2002). However, a recent study indicates the PFA-100® has limited
benefit in evaluating the effectiveness of treatment (Level II) (Favaloro et al. 2005).
j
Including data on all Australian states except NSW
k
FVIII:C= factor VIII coagulant activity
Use of recombinant and plasma-derived Factor VIII and IX
69
While it may detect changes in blood flow after DDAVP treatment, it was not useful in
detecting improvements after treatment with pdFVIII/vWF concentrate.
Table 26.
Methods of testing efficacy of treatments for von Willebrand disease*
Measure
Definition
BT
Bleeding time— time for the bleeding to stop from a superficial incision on the forearm. There are
several different methods that result in different normal ranges. None of these measures of bleeding
time are very sensitive or specific.
Ivy method= less than 5 minutes
Template method= less than 10 minutes
Duke method= less than 3 minutes
PFA-100®
Platelet function analyser—measures blood flow through a capillary device and calculates the time to
capillary occlusion, ie closure time. Measure of closure time is more sensitive than bleeding time, but
it is also non-specific.
FVIII:C
Coagulant activity of FVIII—refers to specific levels/concentration of plasma FVIII
vWF:Ag
von Willebrand factor antigen—immunologic test, measured by ELISA, for the overall level of
vWF:Ag in test plasma; expressed as a percentage of that in pooled, normal plasma (50–200% of
the level in pooled normal plasma, sometimes expressed as 0.5-2.0 U/mL); levels of vWF:Ag are
reduced in classical von Willebrand disease, usually to a similar extent as the reduction in factor VIII
and vWF:Rco
von Willebrand factor-factor VIII binding—measured by ELISA, which tests the ability of von
Willebrand Factor (vWF) to bind factor VIII
Functional assay that reflects the ability of von Willebrand factor to bind collagen; vWF:CB is low in
all types except 2M. This test is used commonly in Australia.
vWF:FVIIIB
vWF:CB
Ristocetin cofactor
tests
Functional assays that reflect the ability of vWF to aggregate normal platelets in the presence of
ristocetin cofactor; commonly used as the initial test for von Willebrand factor activity as it is low in all
but Type 2N von Willebrand disease
vWF:Rco
Uses 3 reagents: multiple dilutions of plasma, standard amount of normal platelets, and standard
amount of ristocetin. Patient platelet aggregation times are compared to a standard curve to
quantitate von Willebrand factor activity as vWF:Rco
RIPA
Ristocetin induced platelet aggregation uses 2 reagents: fresh platelet-rich plasma and 2
concentrations of ristocetin cofactor. While high concentration ristocetin always stimulates
aggregation of normal platelet-rich plasma, low concentration ristocetin reveals any over-response,
such as that seen in Type 2B.
*Methods are described for the purposes of interpreting literature and may not reflect current official tests and abbreviated terms (Kasper,
2004); ELISA= enzyme linked immunosorbent assay
Previous Australian consensus-based guidelines
•
The use of desmopressin (DDAVP), which is preferred over plasma-derived products and is
usually administered by intravenous infusion, in a dose of 0.3 µg/kg of body weight, diluted in
50 mL of 0.9% saline and infused over 30 minutes.
•
Where DDAVP is not likely to be effective, or is contraindicated (i.e. patients with Type 2B von
Willebrand disease), then factor VIII concentrate or purified von Willebrand factor is the treatment
of choice.
•
Cryoprecipitate is not recommended, as it cannot be virally inactivated and, therefore, carries a
risk of viral infection. However, there are some circumstances where the administration of
cryoprecipitate is justified.
See Appendix G for details (Haemophilia Foundation Australia Medical Advisory Panel 2000)
70
Use of recombinant and plasma-derived Factor VIII and IX
What treatments are used?
Recombinant products with von Willebrand factor are still in the research phase.
Therefore, there are two main treatment options for the treatment of bleeding episodes:
desmopressin (DDAVP) or transfusion with plasma-derived products. DDAVP is the
treatment of choice for Type 1 patients, who account for 70–80% of cases (Mannucci
2001). However, DDAVP is contraindicated in Type 2B as it may cause
thrombocytopenia and platelet aggregation due to the release of abnormal platelet
aggregating multimers (Berntorp 1994). DDAVP may also be ineffective in patients with
the more severe Type 3 von Willebrand disease (Level IV evidence) (Schulman et al.
1991). DDAVP should be used with caution in elderly patients and individuals with a
history of heart disease, hypertension or stroke (expert opinion). For more information
on the general recommendations on the use of DDAVP see section on Adjunctive
haemostatic agents.
Patients contraindicated for DDAVP should be treated with a pdFVIII concentrate
containing von Willebrand factor (Mannucci 2001). Biostate® (CSL) is the only
pdFVIII/vWF concentrate for the treatment of von Willebrand disease that is registered
for use in Australia (JBC FVIII/FIX working party & National Blood Authority 2004).
Are treatments effective?
DDAVP
Overall effectiveness of DDAVP for treating bleeding episodes and/or prophylaxis for
patients with von Willebrand disease or mild haemophilia is assessed under the
Adjunctive haemostatic agents section.
Plasma derived factor VIII and von Willebrand factor concentrates
Purified von Willebrand factor concentrates have been in use in France since 1989 (e.g.
Wilfactin®, LFB) for the treatment of von Willebrand disease, but are currently
unavailable in Australia. Therefore, only combined pdFVIII/vWF concentrates like
Biostate® have been assessed for this review.
One randomised crossover trial (Level II evidence) and four case series studies (Level IV
evidence) met our inclusion criteria for assessing the effectiveness of pdFVIII/vWF
concentrates in patients with von Willebrand Disease for bleeding episodes (See Table
27).
The highest level of evidence was a randomised crossover pharmacokinetic study
assessing the effectiveness and safety of two variations of plasma-derived concentrates in
a group of Type 3 von Willebrand disease patients (Level II evidence). This study
reported that Alphanate®, a product not currently available in Australia, reduced median
bleeding time from over 30 minutes to 10.5 minutes. These results suggest that
pdFVIII/vWF concentrates are effective in reducing prolonged bleeding time in patients
with severe von Willebrand disease (Level II evidence) (Mannucci et al. 2002).
Several large case series (Level IV evidence) reported subjective data on the effectiveness
of pdFVIII/vWF concentrates for treating bleeding episodes in von Willebrand patients.
Haemostasis was rated by the clinician as ‘excellent/good’ in the majority of patients
Use of recombinant and plasma-derived Factor VIII and IX
71
after treatment (Level IV evidence) (Lillicrap et al. 2002; Dobrkovska et al. 1998;
Auerswald et al. 2002; Gill et al. 2003).
Dobrkovska et al. (1998) reported that in a group of six patients with prolonged bleeding
time, bleeding times for all patients had normalised at six and 22 hours after infusion
(data not shown). These findings support the recommendation that pdFVIII/vWF
concentrates can be administered every 8–12 hours depending on the individual response
(Level IV evidence) (Dobrkovska et al. 1998).
The retrospective case series by Lillicrap et al. (2002) reported on the efficacy of
Haemate® P in patients where DDAVP was known to be inadequate and where
prophylaxis prior to surgery was indicated. The median dose given to patients to control
bleeding episodes was 55.3 IU vWF:Rco/kg (range 17.1–227.5), the wide range indicating
that individual assessment and monitoring of patients is essential. The minimum,
haemostatically effective dose could not be ascertained from these data (Lillicrap et al.
2002).
72
Use of recombinant and plasma-derived Factor VIII and IX
Table 27.
Effectiveness of plasma-derived factor VIII/von Willebrand factor concentrates in controlling
bleeding episodes
Study
(Mannucci et al.
2002)
Level and quality
of evidence
Population
Level II: randomised
crossover trial
12 patients with Type 3
von Willebrand disease,
treated with Alphanate®
Solvent Detergent (A-SD)
and Alphanate® Solvent
Detergent/Heat-Treated
(A-SD/HT)
Quality score: 22/27
Selection bias
minimised
Confounding
minimised
Effectiveness
Efficacy
FVIII:C
vWF:Ag
vWF:Rco
Mean in vivo half lives (hours)
A-SD
20.9
12.4
7.1
A-SD/HT
23.8
12.9
6.5
Mean in vivo incremental recoveries (per IU/kg)
Follow-up adequate
A-SD
2.0%
2.5%
External validity
uncertainc
A-SD/HT
2.1%
2.9%
Level IV: case series
Quality score: 3/3
Measurement bias
unlikely
Selection bias
minimised
Follow-up adequate
Median bleeding time
14 patients with von
Willebrand disease,
treated with 135 infusions
of Alphanate® for 87
bleeding episodes
Baseline
1 hour post treatment
>30 minutes
10.5 minutes (Range=4.5-20.8
minutes)
All bleeding episodes controlled by treatment, no
patients required alternative therapy
75% of bleeding episodes were controlled after 1
infusion
3 x Type 1
7 x Type 2A
4 x Type 3
Uncontrolled
(Gill et al. 2003)
Level IV:
prospective case
series
Quality score: 3/3
Measurement bias
unlikely
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Auerswald et al.
2002)
Level IV: before and
after case series
Quality score: 3/3
Measurement bias
unlikely
33 patients with von
Willebrand disease,
treated with 290 infusions
of Haemate® P for 53
bleeding episodes—both
surgical and spontaneous
bleeding episodes
52/53 (98%) bleeding episodes had
‘excellent/good’ haemostasis after treatment
9 x Type 1
4 x Type 2A
7 x Type 2B
27 x Type 3
6 x Other
14 patients with vWD,
treated with Immunate®
for 14 acute bleeds or
surgical events
Haemostasis after treatment of bleeding episodes
considered ‘Excellent’ or ‘Good’ in all patients
Perioperative bleeds normal in most cases
3/14 patients experienced post operative
complications, 2 related to insufficient cover
Selection bias
minimised
Follow-up adequate
(cont.)
Uncontrolled
Use of recombinant and plasma-derived Factor VIII and IX
73
Table 27 (cont.)
(Lillicrap et al.
2002;
Dobrkovska et
al. 1998)
Level IV:
retrospective case
series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
97 patients with von
Willebrand disease, 344
bleeding events treated
with Haemate® P
Type 1 vWD: ‘Excellent/good’ haemostasis in 32/32
(100%) bleeding episodes
32 x Type 1
5 x Type 2A
18 x Type 2B
28 x Type 3
14 x Other
Type 2B vWD: ‘Excellent/good’ haemostasis in
60/60 (100%) bleeding episodes
Follow-up adequate
Type 2A vWD: ‘Excellent/good’ haemostasis in
17/17 (100%) bleeding episodes
Type 3 vWD: ‘Excellent/good’ haemostasis in
198/208 (95%) bleeding episodes, ‘Poor’
haemostasis in 10/208 (5%) bleeding episodes
Other types vWD: ‘Excellent/good’ haemostasis in
25/27 (93%) bleeding episodes, ‘Poor’ haemostasis
in 2/27 (7%) bleeding episodes
Uncontrolled
FVIII:C (IU dL-1 per IU kg-1)
Median in vivo recovery= 1.94, Range= 1.21-4.58
vWF:RCo (IU dL-1 per IU kg-1)
Median in vivo recovery= 1.23, Range= 0.59-2.44
(Dobrkovska et
al. 1998)
Level IV: case series
Quality score: 2/3
Measurement bias
unlikely
Selection bias
possible
6 patients with von
Willebrand disease, given
a bolus infusion of
Haemate® P (80 IU
vWF:RCo kg-1 bw and 32
IU FVIII:C kg-1 bw)
Test dose
Follow-up adequate
2 x Type 1B
Uncontrolled
2 x Type 2A
2 x Type 3
(Federici et al.
2002)
Level IV:
retrospective case
series
Quality score: 1/3
10/22 patients with von
Willebrand disease,
treated with Fanhdi®b for
12 bleeding episodes
Measurement bias
possible
9 x Type 1
Unable to assess
selection bias
6 x Type 3
Efficacy
vWF:RCo a
vWF:Ag a
FVIII:C a
In vivo recovery (IU dl/kg)
2.10
(1.10-2.74)
1.88
(1.67-2.90)
2.69
(1.94-3.65)
In vivo recovery (%)
73 (50-96)
69 (51-102)
99 (66-168)
Half-life (hours)
11.3 (6.4-13.3)
15.2 (13.2-17.4)
‘Excellent/good’ haemostasis in 11/12 (92%)
episodes
In 7/12 (58%) cases bleeding episodes were
controlled after one infusion
7 x Type 2B
Follow-up adequate
Uncontrolled
values expressed as median (range); b not available in Australia; c Information on population source not provided; A-SD = Alphanate®
Solvent/detergent treated; A-SD/HT = Alphanate® solvent/detergent, heat-treated; FVIII:C = factor VIII coagulant activity; IVR= in vivo
recovery; RCoF = ristocetin-cofactor activity; vWF:Ag = von Willebrand factor antigen; vWF = von Willebrand factor
a
Are treatments safe?
DDAVP
The safety of DDAVP is discussed under the Adjunctive haemostatic agents section.
Plasma-derived factor VIII and von Willebrand factor concentrates
Recombinant factor VIII concentrates contain no von Willebrand factor and high purity
pdFVIII concentrates contain less von Willebrand factor than intermediate-purity
pdFVIII concentrates. Therefore, although intermediate-purity factor concentrates are
preferred for von Willebrand disease patients, they may increase their risk of contracting
blood borne viruses (Berntorp 1999). The safety issues surrounding treatment with
74
Use of recombinant and plasma-derived Factor VIII and IX
Haemate® Pl or Biostate® are similar to those associated with other intermediate-purity
plasma derived factor concentrates (See Treatment of Bleeding Episodes).
One small case series (Level IV evidence) reported adverse effects in 33.3% of patients
administered Haemate® P (Dobrkovska et al. 1998). Although these side effects were
mild, they occurred after only one administration of factor.
In a larger retrospective case series (Level IV evidence), 17% of patients experienced
adverse events after the administration of Haemate® P (Lillicrap et al. 2002; Dobrkovska
et al. 1998). However, neither study distinguished between patients experiencing these
adverse events in the surgical, bleeding episodes or prophylaxis groups.
Overall, with the exception of isolated cases of deep vein thrombosis, paraesthesia, and
virus transmission (not assessed by the included studies), it appears that most of the side
effects associated with use of pdFVIII/vWF concentrates in von Willebrand disease
were mild in nature (Level IV evidence).
Table 28.
Safety of plasma-derived products for von Willebrand Disease for treating bleeding
episodes
Study
(Mannucci et al.
2002)
Level and quality of
evidence
Population
Level IV: prospective case
series
81 patients with vWD, treated with
Alphanate®-Solvent Detergent (A-SD) or
Alphanate®Solvent Detergent/HeatTreated (A-SD/HT) for 71 surgical or
invasive procedures, or 87 bleeding
episodes
Quality score: 3/3
Measurement bias unlikely
Selection bias minimised
Follow-up adequate
Uncontrolled
(Gill et al. 2003)
Level IV: prospective case
series
Quality of evidence: 3/3
Measurement bias unlikely
Selection bias minimised
Follow-up adequate
Uncontrolled
(Auerswald et al.
2002)
Level IV: case series
Quality of evidence: 3/3
Measurement bias unlikely
15 x Type 1
29 x Type 2A
5 x Type 2B
32 x Type 3
33 patients with von Willebrand disease,
treated with Haemate® P for 53 bleeding
episodes
9 x Type 1
4 x Type 2A
7 x Type 2B
27 x Type 3
6 x Other
14 patients with von Willebrand disease
treated with Immunate® for surgical or
bleeding episodes
Selection bias minimised
Follow-up adequate
Uncontrolled
l
Adverse events
A-SD
A-SD/HT
9/66 (13.6%)
5/36 (13.9%)
Most adverse events were
mild–moderate:
Reduced in vivo recovery,
B19 parvovirus transmission,
thrombophlebitis, deep vein
thrombosis
24/53 (45%) bleeding
episodes associated with
adverse events, but none
considered related to
treatment:
Nausea, vomiting, anaemia,
insomnia
No serious adverse events
One phlebitic reaction at
venous access site after 102
hours of continuous infusion
(cont.)
Haemate® P is not available in Australia
Use of recombinant and plasma-derived Factor VIII and IX
75
Table 28 (cont.)
(Dobrkovska et al.
1998)
Level IV: prospective case
series
Quality score: 2/3
Measurement bias unlikely
Selection bias possible
Follow-up adequate
6 patients with von Willebrand disease,
given a bolus infusion of Haemate® P (80
IU vWF:RCo kg-1 b.w. and 32 IU FVIII:C
kg-1 b.w.)
2/6 (33.3%) patients suffered
mild adverse effects including
urticaria, rash, hives, cough,
fever, weakness and nausea.
2 x Type 1B
2 x Type 2A
2 x Type 3
Uncontrolled
(Lillicrap et al 2002;
Dobrkovska et al.
1998)
Level IV: retrospective
case series
Quality score: 2/3
Measurement bias
possible
Selection bias minimised
Follow-up adequate
Uncontrolled
(Federici et al.
2002)
Level IV: retrospective
case series
Quality score: 1/3
Measurement bias
possible
Unable to assess selection
bias
97 patients with von Willebrand disease,
treated with Haemate® P for 530 different
treatment events: 73 surgery, 344
bleeding events, 20 prophylaxis prior to
surgery, 93 other events
16/97 (16.5%) patients
suffered adverse effects
including chills, phlebitis,
paraesthesia, vasodilation,
pruritis, rash and urticaria.
32 x Type 1
5 x Type 2A
18 x Type 2B
28 x Type 3
14 x Other
10/22 patients with von Willebrand
disease, treated with Fanhdi® for 12
bleeding episodes
No adverse events
9 x Type 1
7 x Type 2B
6 x Type 3
Follow-up adequate
Uncontrolled
A-SD= Alphanate® Solvent/Detergent; A-SD/HT= Alphanate® Solvent Detergent/Heat-Treated
Evidence-based clinical practice guidelines
•
Based on the available evidence, DDAVP may be used in von Willebrand patients who are
responsive to DDAVP and when it is not contraindicated (e.g. DDAVP should not be used during
pregnancy or labour, in children under two years, or in patients with Type 2B von Willebrand
disease) (Level IV evidence).
•
Patients unresponsive to DDAVP or those contraindicated for DDAVP should be treated with
pdFVIII/vWF concentrates (Level IV evidence).
•
Since the optimal dose of Biostate® cannot be determined, close monitoring is required (Level IV
evidence).
•
Plasma-derived FVIII/vWF concentrates may be administered every 8–12 hours depending on the
individual response (Level IV evidence).
Other rare bleeding disorders
There were no studies available that met the predetermined criteria for inclusion in this
review to assess the safety and effectiveness of treatment of bleeding episodes in patients
with other rare bleeding disorders.
76
Use of recombinant and plasma-derived Factor VIII and IX
Prophylaxis
Prophylaxis is defined as ‘treatment by intravenous injection of factor concentrate in
anticipation of and in order to prevent bleeding’ (Berntorp et al. 2003). Instead of simply
using factor concentrates reactively for acute bleeding episodes, a more premeditated
approach of prophylactic administration of factors has been introduced to prevent acute
bleeding episodes and cover surgical procedures. Prophylactic treatment may be further
categorised into primary or secondary prophylaxis. According to the European Network
for Paediatric Haemophilia Management, primary prophylaxis is defined as regular,
continuous treatment started before the age of two years or after the first joint bleed.
Secondary prophylaxis is the regular, continuous (long-term) treatment started at the age
of ≥ two years or after two or more joint bleeds; or, periodic treatment (short-term), due
to frequent bleeds (Petrini 2001).
Young boys with a severe form of haemophilia (A or B) may experience frequent
bleeding into soft tissues (haematomas) or joints (haemarthrosis), leading to joint damage
(arthropathy) and disability by early adult life (Carcao & Aledort 2004). Haemarthrosis
induces acute inflammation and swelling that presents as pain and loss of function. Acute
haemarthrosis may predispose patients to repeated cycles of bleeding in a joint, leading to
chronic joint damage and significant disability. The most widely used measure of
haemophilic arthropathy is the Pettersson radiological score, which classifies
roentgenograms of knees, elbows, and ankles on a scale from 0 to 13 (Pettersson et al.
1981). Scores are given for several changes, which are listed in Table 29. The minimum
score of 0 signifies no arthropathy and the maximum score is 78.
Table 29.
Pettersson radiological score of haemophilic arthropathy
Type of change
Finding
Score
Osteoporosis
Absent
Present
0
1
Enlarged epiphysis
Absent
Present
0
1
Irregular subchondral bone
Absent
Surface partly involved
Surface totally involved
0
1
2
Narrowing of joint space
Absent
Joint space <1mm
Joint space >1mm
0
1
2
Subchondral cyst formation
Absent
1 cyst
>1 cyst
0
1
2
Erosions at joint margins
Absent
Present
0
1
Incongruence of joint surfaces
Absent
Slight
Pronounced
0
1
2
Joint deformity
Absent
Slight
Pronounced
0
1
2
Modified from (Rodriguez-Merchan et al. 2003)
Use of recombinant and plasma-derived Factor VIII and IX
77
Two alternative expanded instruments for measuring arthropathy have been developed in
Colorado—the Colorado Physical Examination full point instrument (Colorado PE-1)
and the Colorado Physical Examination half-point instrument (Colorado PE-0.5)
(Manco-Johnson et al. 2000). These instruments have been designed to detect early
structural and functional abnormalities and to classify the severity of joint impairment. A
third instrument, the Child Physical Examination instrument (Child PE), has been
tailored to reflect the developmental changes in children.
The rationale for prophylaxis in patients with severe haemophilia is underpinned by two
concepts: 1) patients with mild–moderate haemophilia have fewer bleeding episodes with
mild or no arthropathy (Gringeri 2003); and 2) maintaining plasma clotting factor levels
>1% is expected to reduce the number of spontaneous bleeding episodes in patients with
severe haemophilia and prevent secondary arthropathy from recurrent joint
haemorrhages.
The majority of patients undergoing prophylactic treatment are young boys with severe
congenital haemophilia A. Effective prophylaxis depends on sustaining adequate plasma
levels of FVIII to minimise the risk of haemorrhage (Gruppo et al. 2003). Frequent
prophylactic treatment in young children requires adequate access to veins. While regular,
frequent venipuncture may be painful and traumatic in young children, intravenous
access devices may also be associated with complications, such as infections and
thrombosis. For evaluation of the safety and effectiveness of intravenous access devices,
see section on Other Aspects of Management.
Haemophilia
The products used for prophylactic treatment of patients with haemophilia A, without
inhibitors, are the same as those used for treatment of acute bleeding episodes (See
section on Treatment of Bleeding Episodes).
Previous Australian consensus-based guidelines
•
FVIII and FIX prophylaxis is recommended for all children with severe haemophilia A or B up to the
age of 18 years.
•
Haemophilia A: The recommended dosage range for FVIII prophylaxis is 25–40 IU/kg, three times
per week or more frequently as required.
•
Haemophilia B: The recommended dosage range for FIX prophylaxis is 40–60 IU/kg, twice a week
or more frequently as required. See Appendix G for details
(Haemophilia Foundation Australia Medical Advisory Panel 2000).
Are prophylactic treatments effective?
The ‘gold standard’ of prophylaxis is thought to be the Malmö approach, which has been
used in Sweden since 1958 (Carcao & Aledort 2004). The Malmö regimen recommends
prophylaxis in boys with severe haemophilia at 1–2 years of age, before joints are
affected by repeated bleeds, using 25–40 IU/kg pdFVIII every second day (haemophilia
78
Use of recombinant and plasma-derived Factor VIII and IX
A) or pdFIX twice a week for boys with haemophilia B. Follow-up of 60 patients (52
haemophilia A; 8 haemophilia B) over 25 years showed almost no spontaneous bleeds,
regular joints (assessed by Pettersson scores), and good quality of life in boys treated
early (1–2 years), while those given lower doses or treated later showed some
breakthrough bleeding and minor joint defects (Level IV evidence) (Nilsson et al. 1992).
On the basis of these results, the Medical and Scientific Advisory Council (MASAC) of
the National Hemophilia Foundation (USA) recommended prophylactic treatment as
optimal for children with severe haemophilia A or B (Medical and Scientific Advisory
Council (MASAC) 2001).
To date, few good quality randomised controlled trials have assessed prophylaxis in
patients with haemophilia and even fewer have investigated the effectiveness and/or
safety of recombinant factors for prophylaxis.
In this review, several studies on prophylactic treatment were not included for evaluation
as it was impossible to distinguish between patients treated with recombinant and those
treated with plasma-derived factors, or else the type of factor was not stated.
One average quality systematic review of 13 observational studies (Level III-2, III-3 and
IV evidence) compared the effectiveness of full-length FVIII (FL-FVIII), which included
both recombinant and plasma-derived factor, to B-domain-deleted recombinant FVIII
(BDD-rFVIII)m in prophylactic treatment (Gruppo et al. 2003). Meta-analysis showed
that breakthrough bleeding episodesn were more than twice as likely to occur with BDDrFVIII (16.8 bleeds per patient-year [95% CI 9.5-24.2]) than FL-FVIII (6.6 bleeds per
patient-year [4.7-8.5], p<0.0005) at equivalent doses and ages (incidence rate ratio = 2.10
[1.98-2.24]). Pharmacokinetic analysis revealed biochemical differences, such as a shorter
half-life of BDD-rFVIII, which may partly account for the lower prophylactic
effectiveness of BDD-rFVIII. Subsequent analysis of pooled data, using a median-tomean conversion statistico, confirmed the robustness of increased incidence of bleeding
in patients treated with BDD-rFVIII compared to FL-rFVIII (Gruppo et al. 2004).
However, although the investigators used a random effects model to accommodate
heterogeneity when pooling data from the observational studies, these study designs are
prone to selection bias, observer bias and confounding. As such, the results should be
confirmed by well-designed randomised clinical trials that make direct comparisons
between BDD-rFVIII and FL-FVIII groups.
Apart from one good quality randomised crossover trial on prophylaxis in haemophilia B
patients (Level II evidence), current evidence of the effectiveness of prophylactic
treatment is derived primarily from poor–average quality uncontrolled studies (Level IV
BDDrFVIII: The reduced size of the molecule means it is less prone to proteolytic degradation and,
therefore, stabilising with human albumin is not necessary (Fijnvandraat et al. 1997)
m
Pooled incidence of bleeding was calculated from the mean annual total of bleeding episodes reported in
studies
n
o A median-to-mean conversion factor of 2.6 is based on clinical data compiled by the Universal Data
Collection Program of the US Centers for Disease Control and Prevention
Use of recombinant and plasma-derived Factor VIII and IX
79
evidence) that include heterogeneous study populations (haemophilia A or B;
children/adults; PUPs/PTPsp).
The good quality randomised double-blinded crossover trial (Level II evidence)
measured the recovery of plasma FIX following a single infusion of 50 IU/kg rFIX
(BeneFIX®) or pdFIX (Mononine®) in boys with haemophilia B (Kisker et al. 2003).
The aim of this study was to determine the level of dosing necessary to maintain a
prophylactic level of FIX ≥2%. Results are provided in Table 30 and show a twofold
greater recovery with pdFIX compared to rFIX, despite a significantly better median
half-life for rFIX (13.7 vs. 12.9, p=0.016). Pharmacokinetic analysis indicates the median
concentrate required to maintain ≥2% plasma FIX for 30 days, when administered every
third day is 677 IU/kg pdFIX (388-6005 IU/kg/month) compared to 1168 IU/kg rFIX
(268-13085 IU/kg/month). This statistically significant reduction in FIX recovery
following treatment with rFIX also indicated clinical harmq. However, the large overlap
in products, and wide inter-patient variability in recovery and half-life suggests that
individual pharmacokinetic evaluation with different products may be necessary to tailor
appropriate products and dosing schedules to individual needs.
One good quality uncontrolled before-and-after study (Level IV evidence) assessed the
effectiveness of prophylaxis compared with baseline (on-demand treatment) of patients
with severe haemophilia A or B, using recombinant factors VIII or IX (Panicker et al.
2003). While receiving prophylaxis, all patients showed statistically significant reductions
—from baseline—and clinical benefitr in the mean number of bleeding episodes, hospital
admissions, length of hospital stay, and number of emergency room visits (p<0.001)
(Table 30). As expected, there was a concomitant increase in the mean annual dose of
factor when children were on prophylaxis.
p
PUPs = previously untreated patients; PTPs = previously treated patients
Clinical importance of statistically significant effects was ranked according to the checklist provided in
Appendix H. More than 20% reduction in FIX recovery—vs control group FIX recovery—indicated a
clinical harm.
q
r More than 20% improvement (reduction in annual number of major bleeds vs baseline) indicated a
clinical benefit.
80
Use of recombinant and plasma-derived Factor VIII and IX
Table 30.
Factor recovery following prophylactic treatment with recombinant factors in haemophilia A
or B patients
Study
Level and quality
of evidence
(Kisker et al.
2003)
Level II:
randomised
double-blinded
crossover trial
Quality score:
21/27
Clinical
importancea: 1/4
Population
Effectiveness
15 non-bleeding
severe
haemophilia B
patients, aged
>12 years,
without
inhibitors
Incremental factor recovery, IU/kg-1 (range)
rFIX
(50 IU/kg,
BeneFIX®)
pdFIX
(50 IU/kg,
MonoFIX®)
% change
0.86
(0.39-1.48)
1.67
(0.86-4.66)
48.5%
p=0.002
Before prophylaxis
mean±SD (range)
After prophylaxis
mean±SD (range)
Annual number
of major bleeds
15.5±8.3 (6.6-43.5)
1.9±1.2 (0-5.2)
Annual hospital
admissions
1.2±1.5 (0-6.6)
0.1±0.2 (0-0.8)
Annual number
of days in
hospital
7.1±11.6 (0-57.4)
0.4±0.8 (0-2.9)
Annual ER
visits
4.2±3.9 (0-16.2)
0.3±0.4 (0-1.1)
Mean doses
FVIII per year
44.2 (14.1-254.1)
157.6 (105.4-167.6)
Mean doses
FIX per year
17.7 (16.9-18.6)
Selection bias
minimised
Confounding
avoided
Follow-up
adequate
Good external
validity
(Panicker et al.
2003)
Level IV:
uncontrolled
before-and-after
study
Quality score: 2/3
Clinical
importancea: 1/4
Selection bias
minimised
Follow-up
adequate
22 patients with
severe
haemophilia A
and 3 children
with severe
haemophilia B,
aged 1.7–20.4
years, on
prophylaxis
Haemophilia A:
RecombinateTM
or Kogenate®
Haemophilia B*:
BeneFIX®
* 1 patient
received
plasma-derived
FIX
p<0.001
p<0.001
p<0.001
p<0.001
p<0.001
112.6 (108.1-115.4)
p<0.001
Rank scores for the clinical importance of the benefit/harm (1/4 ranked highest and 4/4 ranked lowest); MTPs = minimally treated patients;
PTPs = previously treated patients; PUPs = previously untreated patients
a
Ongoing clinical trials
Three clinical trials of prophylactic treatment are currently ongoing:
ESPRIT (Evaluation Study on Prophylaxis Italian Trial): A prospective randomised trial
(Level II evidence) that compares the efficacy and safety of prophylaxis and on-demand
regimens in reducing the number of bleeding episodes and preventing joint deterioration
in patients with haemophilia A (Gringeri 2003). A one-year follow-up to this study has
been provided in a conference abstract (Gringeri 1998). All patients in the prophylaxis
arm of the study (N=20) maintained FVIII >2%, and no breakthrough bleeding episodes
were reported. One patient developed an infection at the site of the in-dwelling catheter
and one patient, who developed inhibitors to FVIII, was withdrawn from the study. It is
not clear whether the factor concentrates used were plasma-derived or recombinant.
Use of recombinant and plasma-derived Factor VIII and IX
81
US study: A randomised controlled trial (Level II evidence) that aims to determine
whether preventing bleeding into joints is necessary for preventing joint damage in
patients with haemophilia A. This study compares standard prophylaxis (25 IU/kg FVIII
every other day) with an aggressive episode-based regimen for acute joint haemorrhages
(40 IU/kg FVIII at time of haemorrhage onset, 20 IU/kg FVIII at 24 and 72h, and 20
IU/kg FVIII every 48h up to 4 weeks until joint function is completely restored). Thus
far, 65 children from 15 haemophilia centres have been enrolled. The type of FVIII
(plasma-derived or recombinant) is not reported (Manco-Johnson & Blanchette 2003).
Canadian cohort study: A single-arm open-label study (Level III-2 evidence) that
examines whether escalating prophylaxis successfully maintains haemostatic control in
children with severe haemophilia A. All children begin with an initial dose of 50 IU/kg
rFVIII per week. Dosage escalates to 30 IU/kg twice weekly or 25 IU/kg thrice weekly if
a threshold of bleeding episodes (≥3 haemorrhages within 3 months) is exceeded. To
date, 25 children have been followed up for five years, with no losses to follow-up, and
no evidence of safety concerns (Manco-Johnson & Blanchette 2003).
Are prophylactic treatments safe?
Development of inhibitors
The first bleeding episodes in patients with severe haemophilia occur at very different
ages. While evidence suggests that prophylaxis is effective at preventing joint damage,
prophylaxis given before bleeding episodes start is costly and the age of first FVIII
exposure may influence the risk of inhibitor development (Pollmann et al. 1999). Two
retrospective case series found that patient age at initial treatment with factor
concentrates predicted inhibitor development in patients with severe haemophilia (Table
31). Those who were treated before six months of age had a much higher risk of
developing inhibitors than those who were treated between 6–12 months or over 12
months of age. This relationship was not related to cumulative exposure days and no
difference was found between baseline FVIII levels. These results suggest that the risk of
inhibitor development is decreased if the first exposure to factor concentrates is delayed
until there is a clinical need (Level IV evidence) (Lorenzo et al. 2001; van der Bom et al.
2003).
82
Use of recombinant and plasma-derived Factor VIII and IX
Table 31.
Inhibitor development in young children undergoing prophylaxis
Study
Level and quality of
evidence
(Lorenzo
et al.
2001)
Level IV: retrospective
case series
Quality score: 2/3
Selection bias minimised
Population
62 consecutive
severe
haemophilia A
patients
(FVIII<2 IU/dl)
Age at first FVIII exposure
Cumulative
incidence of
inhibitors at 3
years from first
FVIII exposure
0–6
months
7–12
months
>12 months
41%
29%
12%
p=0.03
Follow-up adequate
Uncontrolled
(van der
Bom et al.
2003)
Level IV: retrospective
case series
Quality score: 2/3
Selection bias minimised
81 consecutive
severe
haemophilia A
patients
(FVIII<1 IU/ml)
Follow-up adequate
Uncontrolled
Cumulative
incidence of
inhibitors at 100
days from first
FVIII exposure
4/13 (31%)
Mean number of
infusions before
antibodies
14
IQR=9,44
5/29 (17%)
3/27 (11%)
p=0.03
25
IQR=10,34
40
IQR=10,40
p=0.96
FVIII = factor VIII; IQR= Inter quartile range
Adverse events
One very poor quality historically controlled study (Level III-3 evidence) reported mildsevere adverse events in <1% of haemophilia A patients undergoing prophylaxis (Arkin
et al. 1991).
Table 32.
Adverse events in haemophilia A or B patients undergoing prophylactic treatment with
recombinant factors
Study
(Arkin et al. 1991)
Level and quality of
evidence
Level III-3: historical
control study
Quality score: 5/27
Selection bias likely
Confounding likely
Population
14 non-bleeding PTPs
with moderate–severe
haemophilia A, aged
4–72, without inhibitors
Historical controls: no
description provided
Follow-up uncertain
Adverse events
< 1% of 821 infusions of rFVIII resulted in
adverse reactions
Mild chest twinge, cold hands and feet,
erythema/burning at infusion site, unusual taste
in mouth, dry mouth
Severe dizziness, hypotension, rash, shortness
of breath
External validity
uncertaina
a
Information on population source not provided; PTPs = previously treated patients; rFVIII= recombinant factor VIII
Use of recombinant and plasma-derived Factor VIII and IX
83
Evidence-based clinical practice guidelines
Based on the evidence, it is recommended that prophylactic treatment is available to patients with
severe haemophilia, without inhibitors (Level II and IV evidence):
•
Haemophilia A: Recombinant FVIII—as in consensus guidelines (Level IV evidence).
•
Haemophilia B: Recombinant FIX is safe for prophylaxis in patients with haemophilia B without
inhibitors. Due to the lower rFIX recovery compared to pdFIX, and the large inter-patient
variability, individual dosing regimens should be monitored by FIX recovery/activity assays (Level
II evidence). The recommended rFIX dosage, compared to pdFIX, should be increased by a
multiplication factor of 1.6 for patients aged ≤15 years and 1.2 for patients aged 16 years and
over (Level III-3 evidence, see section on treatment of bleeding episodes).
•
Prophylaxis with factor concentrates should be initiated after bleeding episodes have
commenced, due to an increased risk of inhibitor development when factors are administered at
a young age (Level IV evidence).
84
Use of recombinant and plasma-derived Factor VIII and IX
von Willebrand disease
Previous Australian consensus-based guidelines
There are currently no Australian consensus guidelines for primary prophylaxis in patients with von
Willebrand disease.
Effectiveness of prophylaxis
Although short-term prophylaxis may be given to von Willebrand patients who are
expecting to undergo a serious surgical or dental procedure (see section on Surgical and
Dental Procedures), it is not usually required for long-term management. The exception
to this is the prophylactic treatment of women with menorrhagia, which occurs in over
50% of women with von Willebrand disease (Kadir et al. 2002).
DDAVP and Tranexamic acid
The highest level of evidence on the effectiveness of DDAVP for prophylaxis in von
Willebrand disease was a randomised controlled trial, which found that intranasal
DDAVP was no better than placebo at reducing subjectively measured excessive
bleeding from menorrhagia (Level II evidence) (Kadir et al. 2002).
While there is insufficient evidence to make recommendations based on the effectiveness
of tranexamic acid for treating menorrhagia, two low level studies reported positive
effects in seven patients with von Willebrand disease who were treated for menorrhagia
with a single or daily dose of tranexamic acid (Ong et al. 1998; Mohri 2002). More good
quality research is required in this area before strong recommendations can be made.
Expert opinion suggests that a combination of DDAVP and antifibrinolytics should be
investigated for menorrhagia in von Willebrand disease patients (Kadir et al. 2002).
Plasma-derived factor concentrates
Two low level studies (Level IV evidence) reported on the use of plasma-derived
concentrates for prophylaxis in 20 patients with Type 3 von Willebrand disease—mostly
to prevent recurrent gastrointestinal bleeding. All patients received Haemate® P as
prophylaxis for at least three days and approximately nine patients received prophylaxis
for ≥20 days. A physician efficacy rating of ‘excellent/good’ indicated that there was an
effective reduction in the number of bleeding episodes observed. The median dose for
patients undergoing prophylaxis was 41.6 IU vWF:RCo/kg (range 34.6–81.0). While
there is insufficient evidence for firm recommendations, it appears that prophylaxis with
Haemate® P may be effective for prophylaxis in patients with von Willebrand Disease.
Use of recombinant and plasma-derived Factor VIII and IX
85
Table 33.
Effectiveness of prophylaxis in von Willebrand Disease
Study
(Kadir et al.
2002)
Level and quality of evidence
Level II: randomised crossover trial
Effectiveness
DDAVP nasal spray
Confounding avoided
Median pre-treatment PBAC
score= 250
p=0.51
Level IV: retrospective case series
97 patients with von Willebrand
disease, of which 20 patients
with Type III were given
prophylaxis, mainly to prevent
gastrointestinal bleeding.
Haemate® Pa
4 women with von Willebrand
disease and menorrhagia, after
oral contraceptive pills and
regular dosing of tranexamic acid
had failed to reduce bleeding
High dose tranexamic acid
Quality score: 24/27
Unable to assess external validity
Bias minimised
(Lillicrap et al.
2002;
Dobrkovska et al.
1998)
Population
39 women with mild–moderate
vWD, heterozygous FXIdeficiency, and carriers of
haemophilia, aged 18–60,
treated for menorrhagia
Quality score: 2/3
Measurement bias possible
Selection bias minimised
Median PBAC score= 148
Placebo
Median PBAC score= 140
20/20 (100%) reported a
physician efficacy rating of
‘excellent/good’
Follow-up adequate
(Ong et al. 1998)
Level IV: retrospective case series
Quality score: 1/3
Measurement bias possible
Selection bias possible
(Mohri 2002)
Follow-up adequate
1 x Type 1
2 x Type 2A
1 x Type 2B
Level IV: retrospective case series
3 women with von Willebrand
disease and menorrhagia, after
DDAVP or pdFVIII/vWF
concentrates had minimal effect
Quality score: 1/3
Measurement bias possible
Selection bias possible
Follow-up adequate
4/4 (100%) had menorrhagia
controlled
High dose tranexamic acid
3/3 (100%) had menorrhagia
well controlled
1 x Type 1
2 x Type 2A
Haemate® P is not available in Australia; FXI= factor XI; PBAC= Pictorial Blood Assessment Chart, a commonly used tool to measure
menstrual loss where a score of ≥100 is equivalent to more than 80 mL of blood loss (definition of menorrhagia); vWD= von Willebrand disease
a
Safety of prophylaxis
The safety of all three of the prophylactic treatments - DDAVP, tranexamic acid and
pdFVIII/vWF concentrates - was assessed in other sections (Adjunctive haemostatic
agents; Treatment of Bleeding Episodes and Surgical and Dental Procedures). The same
safety issues apply for this population and for this purpose.
Evidence-based clinical practice guidelines
Based on limited published evidence, DDAVP administered intranasally is ineffective as prophylaxis to
prevent menorrhagia in women with von Willebrand disease (Level II evidence). Plasma-derived
FVIII/vWF concentrates or tranexamic acid are possible alternatives (Level IV evidence).
Other rare bleeding disorders
There were no studies that met the predetermined criteria for inclusion in this review to
assess effectiveness and safety of prophylaxis in patients with other rare bleeding
disorders.
86
Use of recombinant and plasma-derived Factor VIII and IX
Inhibitors and Tolerisation
With the advent of donor screening and sophisticated processes to inactivate viruses,
such as HIV and hepatitis, and eliminate prions and parvovirus (e.g. Creutzfeld-Jakob
disease; parvovirus B19) with nanofiltration techniques, blood coagulation products of
increasing purity have become available and the risk of transmission of viruses has been
almost eliminated. However, the development of inhibitory antibodies to FVIII or FIX
remains one of the greatest challenges to physicians treating patients with FVIII or FIX
deficiency, particularly in those with severe haemophilia. Therapeutically administered
factor is recognised as a foreign protein by the immune system, thus stimulating the
production of antibodies (inhibitors). Inhibitors react with and neutralise FVIII
rendering it ineffective in the clotting process. If the inhibitor titre is high, factor
replacement therapy is totally ineffective and bleeding persists. With low titre inhibitor
haemostasis may still be achieved, though higher doses of factor concentrates may be
needed. Prophylaxis is not an option for high-titre inhibitor patients and those with
severe haemophilia and high-titre inhibitors are most at risk for recurrent bleeds and
chronic haemarthroses.
The development of inhibitors to FVIII or FIX occurs: 1) in patients with congenital
haemophilia A or B who develop alloantibodies when treated with FVIII or FIX
concentrates; or, more rarely 2) in patients with normal haemostasis who develop
autoantibodies to FVIII or FIX (‘acquired haemophilia’). While the development of
inhibitors to FIX in haemophilia B is relatively rare (1–6%) and usually associated with a
gross gene deletion or nonsense mutation, FVIII inhibitor formation in haemophilia A
patients is more common (up to 33%) (World Health Organization 2002). The
prevalence of inhibitors to FVIII in haemophilia A patients varies widely (4–20% in
moderate-severe patients; up to 50% in study populations with severe haemophilia)
(Manno 1999) depending on the frequency of inhibitor measurement, the study
population (age, ethnicity, immune status), the product used and methods of data
collection. According to the Australian Bleeding Disorder Registry, approximately 8% of
registered patients have inhibitors (Australian Bleeding Disorder Registry 2004).
In patients with severe haemophilia A, inhibitors generally develop after a median of 9–
12 days of exposure to treatment (Hay et al. 2000b). Screening for inhibitors may be
conducted by two methods: activated partial thromboplastin time (aPTT) assays or the
Bethesda method, which is more sensitive and routinely used in Australia. Inhibitor titre
is measured in Bethesda Units (BU)t—a measure of the extent to which plasma with
inhibitor inactivates FVIII in normal human plasma (Paisley et al. 2003). A similar
Bethesda method is used to quantify inhibitors to FIX in haemophilia B patients.
The level of inhibitors varies not only between individuals but also within individuals
over time. Inhibitors are measured on two dimensions: 1) the concentration of
inhibitor—low titre (≤5 BU) and high titre (>5 BU); and 2) the response to factor—low
Activated partial thromboplastin time (aPTT) = detects the presence of substances in the blood that
inhibit the activity of coagulation factors
s
Bethesda units (BU) = a measure of inhibitor activity—the amount of inhibitor that inactivates 50% or
0.5 units of a coagulation factor during the incubation period
t
Use of recombinant and plasma-derived Factor VIII and IX
87
responders show no increase in inhibitors, while high responders show a large increase in
inhibitor production (anamnestic response). Clinically, low responders can be treated
with FVIII concentrate, usually at high doses, whereas high responders cannot.
In most cases the antibody is at a low level (<5 BU/ml) and tends to disappear
spontaneously. However, for approximately 20% of patients, the inhibitor persists,
making prophylaxis and routine treatment with factor concentrates ineffective, which
increases the risks associated with surgical or dental procedures.
Although the frequency and site of bleeding episodes in haemophilia patients is similar
whether or not inhibitors are present, management of the condition is more challenging
and requires different treatment strategies. In general, ad hoc treatment for acute
bleeding episodes in patients with inhibitors requires the use of the alternative therapies
listed in Table 34. Additionally, procedures have been developed to eliminate inhibitors.
These include immune tolerance induction (tolerisation) or immune suppression therapy,
which is used mainly in acquired haemophilia. Evidence on the safety and effectiveness
of tolerisation protocols is addressed in the following section.
88
Use of recombinant and plasma-derived Factor VIII and IX
Table 34.
Products used for haemophilia patients with inhibitors
Type of
product
rFVIII
Mechanism of
action
FVIII replacement
Product
(Manufacturer)
Recombinate™
(Baxter Hyland
Immuno)a
Kogenate® (Bayer)a
BioclateTM (Aventis
Behring)a
Helixate® (Aventis
Behring)a
ReFacto® (Genetics
Institute)
BeneFIX® (Genetics
Institute)
Disadvantages
rFIX
FIX replacement
rFVIIa
Bypassing agent
NovoSeven®
(NovoNordisk)
Plasmaderived FVIII
FVIII replacement
Hemophil® M (Baxter
Hyland Immuno)
Pooled plasma product
Unknown risk of viral
transmission
Plasmaderived FIX
FIX replacement
Alphanine® (Alpha)
MonoFIX (Aventis
Behring)
aPCCs (with
FIIa, FVIIa,
FXa)
Bypassing agent
Autoplex® T (Nabi)
FEIBA VH® (Baxter
Hyland Immuno)
PCCs (with FII,
FVII, FIX)
Bypassing agent
Prothrombinex®-HT
Proplex T; Bebulin VH
(Baxter Hyland
Immuno)
Konyne 80 (Bayer)
Profilnine SD (Alpha)
Pooled plasma product
Anaphylaxis and nephritic
syndrome reported when
used for tolerisation
Unknown risk of viral
transmission
Pooled plasma product
Risk of thrombosis
Anamnestic response
Allergic reaction
Cannot be assayed
Pooled plasma product
Less effective than aPCCs
Risk of thrombosis
Factor content variable
between lots and products
Anaphylaxis and nephritic
syndrome reported when
used for tolerisation
Cannot be assayed
Advantages
Not effective for high titre
inhibitors
Anamnestic response
No known risk of viral
transmission
Measured by laboratory
assay
Used for maintenance and
prophylaxis after successful
tolerisation
Not effective for acute
bleeding episodes in high
titre inhibitors
Short half-life
Thrombosis risk
As for rFVIII
Devoid of human serum or
proteins
No known risk of viral
transmission
No human serum or
proteins
No anamnestic response
Measured by laboratory
assay
Used for maintenance and
prophylaxis after successful
tolerisation
Can be measured by
laboratory assay
Long half-life
Virally inactivated
More effective than PCCs
Long half-life
Virally inactivated
Modified from (Kulkarni et al. 2001); a Product contains albumin; b Due to a series of corporate changes since this reference article was
published in 2001, some product manufacturers in this table may differ from those listed previously in the section under Selection of Products;
aPCCs = activated prothrombin complex concentrates; FEIBA= factor eight inhibitor bypassing agent; PCC = prothrombin complex
concentrates
Use of recombinant and plasma-derived Factor VIII and IX
89
Previous Australian consensus-based guidelines
These guidelines are for the treatment of haemophilia A in patients with inhibitors to FVIII. There are no
existing guidelines for the treatment of haemophilia B in patients with inhibitors to FIX.
Low titre inhibitor (<5 BU/ml), low responder
Minor bleeding
FVIII (50–100 IU/kg every 8–12 hours). Assess clinically and monitor FVIII levels.
Major bleeding
FVIII (50–150 IU/kg every 8–12 hours). Assess clinically. Monitor FVIII levels and maintain at >50% until
healing has completed.
Low titre inhibitor (<5 BU/ml), high responder
Minor bleeding
Recombinant FVIIa (90 µg/kg—adult dose; up to 200–250 µg/kg—paediatric dose) every 2 hours. Assess
clinically and follow up with a further single dose. If no response is observed, consider treatment with aPCC.
Alternative treatment with FEIBA: 60–80 IU/kg FEIBA bd (max = 200 IU/kg/day)
Major bleeding
Recombinant FVIIa (90 µg/kg—adult dose; up to 200–250 µg/kg—paediatric dose) every 2 hours for
minimum 2 doses. Assess clinically. If no response is observed, consider treatment with aPCC.
Alternative treatment with FEIBA: 60–100 IU/kg FEIBA as a single dose
High titre inhibitor (>5 BU/ml)
Minor bleeding
Recombinant FVIIa (90 µg/kg—adult dose; up to 200–250 µg/kg—paediatric dose) every 2 hours for
minimum 2 doses. Assess clinically and follow up with a further single dose. If no response is observed,
consider treatment with aPCC.
Alternative treatment with FEIBA: 60–80 IU/kg FEIBA as a single dose
Major bleeding
Recombinant FVIIa (90 µg/kg—adult dose; up to 200–250 µg/kg—paediatric dose) every 2 hours for 12
hours; increase to 3 hourly intervals depending on clinical response. Increase dose interval to 4 hours if
necessary to maintain dosage. Duration may extend to 14 days if major surgery has been performed.
Alternative dosing regimen: 320 µg/kg every 6 hours. If no response is observed, consider treatment with
aPCC.
Alternative treatment with FEIBA: 60–100 IU/kg FEIBA bd (max = 200 IU/kg/day).
* Modified from guidelines by AHCDO (2004) See Appendix G; aPCCs = activated prothrombin complex concentrates; bd = twice per day;
FEIBA = Factor Eight Inhibitor Bypassing Agent
Treatment of bleeding episodes in patients with inhibitors
The highest level of evidence was provided by one good quality systematic review of 52
studies (Lloyd Jones et al. 2003). Overall, the study design, therapy and methodology of
included studies, which were predominantly low level evidence (Level IV), were highly
variable and of poor quality. Few randomised controlled trials with appropriate
comparators or placebo were available (8 of 52). The target population in the included
studies was predominantly patients with inhibitors to FVIII (haemophilia A), although
some studies included those with inhibitors to FIX (haemophilia B). A wide range of
therapies was used in haemophilia patients with inhibitors, including: 1) high-dose human
FVIII concentrate (plasma-derived); 2) highly purified porcine FVIIIu; 3) inhibitor
bypassing agents—activated prothrombin complex concentrates (aPCCs) and
prothrombin complex concentrates (PCCs); 4) plasmapheresis or immunoadsorption
plus high-dose FVIII; 5) cimetidine plus high-dose FVIII; and 6) immunosuppression
plus plasma-derived FVIII. Data from this systematic review are summarised in Table 35.
Due to the discontinuation of a poor quality randomised controlled trial and the absence
of data from case series, there was no available evidence to support the use of high-dose
plasma-derived FVIII alone for controlling acute bleeding in patients with high titre
u
Since porcine FVIII is not used in Australia, data has not been presented in this report
90
Use of recombinant and plasma-derived Factor VIII and IX
inhibitors or in high responder patients. There was no evidence assessing the safety or
effectiveness of rFVIII for treating bleeding episodes in patients with inhibitors,
however, due to reports of equivalent effectiveness between rFVIII and pdFVIII in
patients without inhibitors, and the higher safety profile of rFVIII, it is suggested that
rFVIII may be a better alternative (Lloyd Jones et al. 2003) (expert opinion). Higher-level
evidence (Level II) indicates that PCCsv and aPCCs are effective in controlling mild to
severe bleeding in patients with inhibitors (≥4 BU/ml). However these inhibitorbypassing agents may be unpredictable, with a potential for serious adverse reactions—
including thrombosis, disseminated intravascular coagulation (DIC), myocardial
infarction and pulmonary embolism. Therefore, they are not recommended for first-line
treatment in life-threatening haemorrhages. In addition, the maximum daily dose of
FEIBA should not exceed 200 IU/kg (Level II evidence, Lloyd Jones et al. 2003).
In terms of both safety and efficacy, the evidence (Level II) suggests that rFVIIa is the
best option as the first line of treatment for high responder patients, particularly if used
early rather than late. Controlling 70–90% of mild–severe bleeding episodes in high
responder patients, rFVIIa does not provoke anamnesis, side effects are generally minor,
and it is suitable for home treatment (Lloyd Jones et al. 2003). Evidence from a
systematic review by Lloyd Jones suggests that acute haemorrhages of mild to moderate
severity are best treated using 90 µg/kg of rFVIIa (Level IV evidence). While the
standard method of rFVIIa administration is by bolus injection, low level evidence
suggests that continuous infusion is also effective (Level IV evidence) (Lloyd Jones et al.
2003). There is evidence that the half life of rFVIIa is shorter in children than in adults
(Level IV evidence, Lloyd Jones et al. 2003). Therefore, the recommendation from
previous consensus guidelines, that children may require as much as 200-250 µg/kg
rFVIIa, should remain.
Limited low-level evidence (Level IV) shows successful reduction of inhibitors by
plasmapheresis (with or without immunoadsorption) and subsequent infusion of
pdFVIII to control bleeding in patients with high titre or high responder inhibitors (80–
100%). In contrast, immunosuppression with cyclophosphamide to prevent anamnesis
combined with factor replacement is largely unsuccessful.
v The formulation of PCCs tested in the Level II evidence is different to what is currently available in
Australia; therefore, while results are presented, recommendations on the use of PCCs have not been given
Use of recombinant and plasma-derived Factor VIII and IX
91
Table 35.
Summary of efficacy of interventions to control bleeding in haemophilia A patients with
inhibitors*
Intervention
Population
Level of evidence
Acute bleeding episodes
pdFVIII
Any
PCCs
High titre/ high
responders
aPCCs
High titre/ high
responders
rFVIIa
High
responders or
high titre
Intermediatehigh responders
High titre
Plasmapheresis + highdose pdFVIII
Extracorporeal protein
A adsorption + highdose pdFVIII
Cimetidine + FVIII
No evidencea
Level II: RCT
Level IV: uncontrolled beforeand-after study
Level II: RCT
Level IV: uncontrolled beforeand-after studies
Level II: RCT
Level IV: uncontrolled beforeand-after studies
Level IV: case series
Haemostasis
control (%)
No evidence
83%
81%
Adverse events
No evidence
Mainly minor
Potential for DIC, DVT, MI
64%
88–100%
71–90%
62–100%
Mainly minor. Occasional
thrombophlebitis or DIC
100%
Level IV: uncontrolled beforeand-after study
80%
Anaphylactic reactions,
headache, fever, vomiting
All minor
High
responders
Any
Level IV: uncontrolled beforeand-after study
Level IV: uncontrolled beforeand-after study
100%
None reported
27%
Increased susceptibility to
infection, impaired wound
healing
Level IV: case series
78%
None reported
aPCCs
High titre/ high
responders
Not clear
86–93%
rFVIIa
High titre
Mainly minor. Occasional
thrombophlebitis or DIC
Mainly minor. Occasional
thrombophlebitis or DIC
Plasmapheresis + highdose pdFVIII
Dental surgery
PCCs
High titre
Level IV: uncontrolled beforeand-after study
Level II: RCT
Level IV: uncontrolled beforeand-after studies
Level IV: case series
67%
Anaphylactic reactions,
headache, fever, vomiting
Level IV: case series
33%
Level IV: case series
100%
Level IV: uncontrolled beforeand-after study; case series
86–92%
Mainly minor. Potential for
DIC, DVT, MI
Mainly minor. Potential for
DIC, DVT, MI
Mainly minor. Occasional
thrombophlebitis or DIC
Immunosuppression +
pdFVIII
Surgical procedures
pdFVIII
aPCCs
High and low
titre
High titre
rFVIIa
Not stated
55–100%
*Modified from (Lloyd Jones et al. 2003) a One poor quality RCT was discontinued (non-compliance) and a case series failed to provide data for
effectiveness; aPCCs = activated prothrombin complex concentrates; DIC = disseminated intravascular coagulation; DVT = deep vein
thrombosis; MI = myocardial infarction; PCCs = prothrombin complex concentrates, (a different formulation than is currently available in
Australia); RCT = randomised controlled trial; rFVIIa= activated recombinant factor VII, pdFVIII = plasma-derived factor VIII
Surgical and dental procedures in haemophilia patients with inhibitors
The best evidence for the management of patients with inhibitors during surgery or
dental procedures is also provided in the systematic review by Lloyd Jones et al. 2003
(Table 35). In surgery, high-dose plasma-derived FVIII was successful for patients with
low-titre, low-responding inhibitors (100%). It was less reliable for controlling peri- and
post-operative bleeding in patients with high-responding inhibitors (80%); only effective
when the inhibitor titre was below 10 BU/ml and only for 5–7 days before an
anamnestic response occurred. Bypassing agents (PCCs, aPCCs, and rFVIIa) are the only
available options for patients who cannot be treated with plasma-derived FVIII. Lowlevel evidence indicates that PCCs, even when combined with antifibrinolytic agents,
have limited success in controlling bleeding associated with dental surgery (33%), with
variable results on different occasions in the same patient. In contrast, aPCCs were
92
Use of recombinant and plasma-derived Factor VIII and IX
successful in controlling bleeding in approximately 90% of surgical and 100% of dental
procedures (with antifibrinolytic agents). However, although most side effects were
minor, there is the potential for serious complications and evidence suggests that
antifibrinolytic products should be used consecutively with aPCCs rather than
simultaneously during dental surgery. The highest level of evidence for use of rFVIIa in
surgery is derived from one randomised controlled trial (Level II evidence), which
demonstrated 93% success. The majority of remaining studies included patients enrolled
in a ‘compassionate use’ programw, which comprises patients who have not or are
unlikely to respond to other treatments. In most cases, their clinical condition is
complicated by disordered haemostasis, advanced age, or general poor health.
Evidence-based clinical practice guidelines
Based on the evidence, it is recommended that haemophilia A patients with inhibitors be treated in the
following manner:
•
Recombinant FVIIa may be considered the first line of treatment for acute bleeding episodes or
dental and other surgical procedures in patients with high titre and/or high responder inhibitors
(Level II evidence).
•
aPCCs may be used to control mild to severe bleeding in patients with high titre inhibitors. Due to
their unpredictable effects, patients should be closely monitored for severe reactions, such as
disseminated intravascular coagulation, myocardial infarction, or thrombotic complications.
Activated PCCs are preferred over PCCs, particularly for controlling bleeding in dental and other
surgical procedures. During dental surgery, aPCCs should be used consecutively with
antifibrinolytic agents, rather than in combination (Level IV evidence). The maximum daily dose of
FEIBA should not exceed 200 IU/kg (Level II evidence).
•
Plasmapheresis (with or without immunoadsorption) may be considered to reduce inhibitors in high
titre/high responders prior to infusion with FVIII concentrate to control bleeding. However, patients
should be closely monitored for anaphylactic reactions (Level IV evidence).
•
Plasma-derived FVIII concentrate may be considered for use during surgery in patients with low
titre, low-responding inhibitors, but should be avoided in patients with high-responding inhibitors
(Level IV evidence). There was no evidence to support its use for the treatment of acute bleeding
episodes. The evidence on recombinant FVIII concentrate is awaited.
•
Immunosuppression therapy with cyclophosphamide to reduce inhibitors should be avoided (Level
IV evidence).
Tolerisation protocols
For the most part, if inhibitors develop they are most likely to do so within a few
exposure days (median 9–12 days) in young boys with severe haemophilia A. In addition,
most low titre inhibitors disappear over time. For a small proportion, however, inhibitors
persist and strategies for eradication of inhibitors need to be employed.
w
Compassionate use program—manufacturer allows physicians to use rVIIa on patients with special needs
Use of recombinant and plasma-derived Factor VIII and IX
93
Currently, the only method of removing inhibitors in patients with haemophilia A or B is
by immune tolerance induction (DiMichele 1998). Several different methods of immune
tolerance induction (tolerisation) have attempted to abolish inhibitors with varying
success, but there is no consensus on the optimum approach of tolerisation.
In Australia, tolerisation protocols are highly variable across haemophilia centres, with no
consensus on procedures. Dosages of FVIII concentrate for tolerisation of haemophilia
A patients with inhibitors to FVIII range from 25 IU/kg three times a week to 100
IU/kg/day (JBC FVIII/FIX working party & National Blood Authority 2004). Since the
current Australian consensus-based guidelines are recently developed, there is no
available evidence of the effectiveness of this approach.
Previous Australian consensus-based guidelines
Tolerisation should be considered in all those patients with recent persisting inhibitors. Written informed
consent should be obtained before starting. Intensive replacement therapy for immune tolerance usually
requires central venous access.
Haemophilia A
•
The International Registry on Tolerisation identifies better results in those patients with lower age
at the start of Immune Tolerance Induction (ITI); shorter elapsed time of inhibitor presence before
ITI; lower maximum pre-treatment inhibitor titres and treatment with higher doses of FVIII.
•
AHCDO strongly recommends participation in an international randomised trial of high dose versus
low dose factor VIII tolerisation protocols. All patients considered eligible for the trial should be
involved.
•
For patients who do not enter an international trial, there are a number of published tolerisation
protocols that describe a variety of doses of factor VIII (e.g. 50 IU/kg three times a week up to 200
IU/kg daily) as well as the use of immune suppression. Tolerisation should continue until
eradication of the inhibitor demonstrated by a greater than 60% recovery and normal half-life of
factor VIII. Tolerisation should only be attempted in consultation with a physician experienced in
the management of patients with haemophilia and inhibitors.
•
It is recommended that adults with severe haemophilia A who have had an inhibitor for many
years, and in some instances decades, should be managed with treatment of haemorrhage by
infusion of recombinant factor VIIa. Adult patients with mild or moderate haemophilia A who
develop inhibitors often have mild inhibitors, which may decline with time. These patients should be
treated with desmopressin (DDAVP) for minor bleeds and recombinant factor VIIa for more serious
haemorrhages. Tolerisation may be considered if haemorrhages cannot be controlled by
recombinant factor VIIa.
Acquired Haemophilia A
Immunosuppression is recommended for eradication of inhibitors in Acquired Haemophilia A.
Haemophilia B
There are no existing Australian clinical practice guidelines for ITI in patients with inhibitors to factor IX.
See Appendix G for details (Australian Haemophilia Centre Directors' Organisation 2004).
94
Use of recombinant and plasma-derived Factor VIII and IX
Effectiveness of tolerisation
The best information on the effectiveness and safety of tolerisation protocols for
haemophilia A with inhibitors comes from one good quality systematic review of level IV
evidence (Wight et al. 2003). No studies of tolerisation in patients with haemophilia B,
von Willebrand disease or other rare bleeding disorders satisfied our inclusion criteria for
an evaluation of effectiveness.
High dose protocols
Bonn protocol: The original Bonn regime involved giving 100 IU/kg FVIII plus 40–60
IU/kg FEIBA every 12 hours until the inhibitor level dropped below 1 BU and FEIBA
was discontinued. Then FVIII was reduced to 150 IU/kg daily until inhibitor was no
longer detectable. Recently, the protocol was modified so that people with a tendency to
bleed are given 150 IU/kg FVIII every 12 hours, rather than FEIBA (Wight et al. 2003).
After completing the Bonn protocol or other high dose protocols, 90% of patients had
complete elimination of inhibitors and partial success in two others (Level IV evidence)
(Wight et al. 2003). In a large case series (Level IV evidence) it was found that lower
initial inhibitor titres were associated with shorter treatment time on the Bonn protocol
(Wight et al. 2003).
Malmö protocol: The Malmö protocol utilises extracorporeal immunoadsorption when
inhibitor titres are above 10 BU, until levels are brought down to less than 10 BU. High
doses of FVIII (around 200 IU/kg/day) are then administered to bring FVIII
concentration up to 40–100 IU/dL and to maintain it at 30–80 IU/dL. At the same time,
cyclophosphamide (an immunosuppressant) is administered at a dose of 12–15 mg/kg
intravenously for two days, followed by 2–3 mg/kg for eight days. On day one of
treatment, immunoglobulin is also used at a dose of 2.5–5 g/kg, followed by 0.4 g/kg on
days 4–8. Factor VIII is administered until the inhibitor disappears and patients receive
FVIII for ongoing prophylaxis. Of the 32 patients who were treated by the Malmö
protocol or slight variants of it, 47% successfully had inhibitors removed (Wight et al.
2003). One case series (Level IV evidence) found that treatment of young children
tended to be unsuccessful with the Malmö protocol, and success was more likely in
patients with long standing inhibitors who had been exposed to multiple doses of FVIII
since inhibitor development (Wight et al. 2003). These results are contrary to those found
in the Bonn protocol, which suggests that while the Malmö protocol may be less
effective overall, it may be more effective than other protocols in patients with long
standing inhibitors (Wight et al. 2003).
The Bonn and Malmö protocols are not used routinely in Australian practice. In
particular, the Malmö protocol requires specific equipment not readily available outside
Sweden and is, consequently, not practical for use in Australia (JBC FVIII/FIX working
party & National Blood Authority 2004).
Other protocols have been developed, aiming to reduce the cost and complexity of the
Bonn and Malmö protocols (Wight et al. 2003). However, there is insufficient evidence
relating to these variations for adequate evaluation.
Low dose protocols
A variety of low dose protocols have also been developed for use in haemophilia
patients. Dose regimens using FVIII varied from 175 IU/kg/week to 100 IU/kg/day,
Use of recombinant and plasma-derived Factor VIII and IX
95
with or without additional immunosuppressive drugs (Wight et al. 2003). For example,
from a total of 107 patients following a low dose protocol (maximum of 100 IU/kg/day
of factor VIII), immune tolerance was successful in 67% of patients, and partially
successful in 8% of others (Level IV evidence).
Low dose regimes may be more cost-effective and may be administered more readily
without the use of a central line (Level IV evidence) (Hay et al. 2000a).
Registry data
The International, North American, German and Spanish registries on haemophilia
patients with inhibitors were analysed in univariate and multivariate analysis. The results
suggest that tolerisation is more likely to be successful when patients are younger,
maximum pre-treatment inhibitor titres are low, and when there is a shorter period of
time between development of inhibitor and treatment of inhibitors (Level IV evidence)
(Wight et al. 2003).
The International Immune Tolerance Study—ongoing
A randomised controlled trial (Level II evidence) of the effectiveness, morbidity and cost
effectiveness of two types of immune tolerance induction in severe haemophilia A was
started late in 2002 and aims to include 150 patients. The study was delayed due to a
shortage of rFVIII and no results have been published to date. Based on the lack of
evidence regarding optimum treatment for eradicating inhibitors, it is suggested that
patients with severe haemophilia A, an inhibitor for 12 months or less, a historical peak
inhibitor titre of > 5 but < 200 BU, and currently an inhibitor titre of less than 10 BU be
enrolled in the International Immune Tolerance Study so future treatment may be guided
by clinical evidence (Hay et al. 2000a). The International Immune Tolerance Study is
expected to be completed by July 2007.
Immunosuppressive drugs
Based on poor quality studies, immunosuppressive drugs used to date are ineffective in
reducing inhibitors in haemophilia patients (Level IV evidence). A survey on the use of
immunosuppressive drugs in 56 patients found that the drugs were ineffective in the
majority of cases (61%), partially effective in 28% of patients and successful in only 13%
of patients (Lloyd Jones et al. 2003). It is possible that newer immunosuppressive drugs,
such as rituximab, may be more effective than those used previously, but there is no
evidence available at present to recommend this treatment.
Choice of products
There is no strong evidence that any particular type or brand of product is more effective
than another in immune tolerance induction. An uncontrolled study on a small group of
people found that an intermediate-purity factor VIII concentrate was more effective than
high-purity factor. However, similar response rates have been reported with high-purity
or recombinant products (Hay et al. 2000a). Therefore, it is recommended that rFVIII
products be used due to the higher safety profile compared with plasma-derived factor.
96
Use of recombinant and plasma-derived Factor VIII and IX
Safety of tolerisation
No available studies reported on the safety of immune tolerance protocols, although the
International Immune Tolerance Study is currently collecting information on adverse
events related to the tolerisation and data should be available in the future.
If tolerisation is performed on patients with haemophilia B using FIX, patients should be
closely monitored to prevent anaphylactic reactions (expert opinion, AHCDO Executive
Committee 2005).
Evidence-based clinical practice guidelines
In Australia, the tolerisation protocols vary widely across haemophilia treatment centres, with little
published evidence of effectiveness, no consensus on protocols, and the Bonn and Malmö protocols are
not routinely used. Doses of FVIII concentrate for tolerisation range from 25 IU/kg three times per week,
to 100 IU/kg/day. Guidelines should be revised after publication of results from the International Immune
Tolerance Study.
•
The Bonn protocol may be considered for young patients (Level IV evidence).
Bonn protocol (modified): People with a tendency to bleed are given 150 IU/kg FVIII every 12 hours until the
inhibitor level drops below 1 BU. Then FVIII is reduced to 150 IU/kg daily, until inhibitor is no longer
detectable.
•
Treatment is more likely to be successful with lower pre-treatment inhibitor titres and less time
between development and treatment of inhibitors (Level IV evidence).
•
When patients have long standing inhibitors, the Malmö protocol may be considered (Level IV
evidence).
Malmö protocol: When inhibitor titres ≥10 BU extracorporeal immunoadsorption is given until levels are
reduced to <10 BU. High doses of FVIII (around 200 IU/kg/day) are then administered to bring FVIII
concentration up to 40–100 IU/dL and to maintain it at 30–80 IU/dL. At the same time, cyclophosphamide is
administered at a dose of 12–15 mg/kg intravenously for two days, followed by 2–3 mg/kg for eight days. On
day one of treatment, immunoglobulin is also used at a dose of 2.5–5 g/kg, followed by 0.4 g/kg on days 4–
8. Factor VIII is administered until the inhibitor disappears and patients receive FVIII for ongoing prophylaxis.
•
When factor concentrates are used, recombinant factors are preferred rather than plasma-derived
factors, due to their higher safety profile regarding transmission of blood-borne agents (Level IV
evidence).
•
There is no evidence to guide tolerisation procedures in patients with haemophilia B with inhibitors.
Recombinant FIX is preferred over plasma-derived FIX due to the higher safety profile pertaining to
transmission of blood-borne agents, but there should be close monitoring to prevent anaphylactic
reactions (expert opinion).
Use of recombinant and plasma-derived Factor VIII and IX
97
Treatment of patients with acquired haemophilia
Acquired haemophilia is a very rare disease (about one in a million people) in which
people without haemophilia develop antibodies to factor VIII. The bleeding
complications associated with acquired haemophilia can be life threatening, so prompt
diagnosis and treatment are vital. While most people with acquired haemophilia have
idiopathic autoantibodies, 40–50% of cases are associated with other conditions, such as
the postpartum state, autoimmune disease, underlying malignancies and drug
administration (Delgado et al. 2003). Due to the rarity of acquired haemophilia,
information regarding its treatment and prognostic factors is derived solely from case
series (Level IV evidence) or from an average quality systematic review of case series
with five or more patients (Level IV) (Delgado et al. 2003). Due to the lack of
appropriate studies for evaluation of effectiveness recommendations must rely on case
series.
Pregnancy
Antibodies to factor VIII usually develop 1–4 months after infant delivery, but some
patients show inhibitors during pregnancy or up to one year after delivery. In the
majority of cases, the level of postpartum inhibitors is low and they disappear
spontaneously after a mean of 30 months (Level IV evidence). However, it is advisable
that patients are evaluated for lupus or rheumatoid arthritis since patients with these
disorders require a different approach to treatment (Delgado et al. 2003).
An important issue in managing pregnancy-related acquired haemophilia is the risk of
inhibitor recurrence in subsequent pregnancies. While most case series reported little risk
in those who achieved complete remission, one study, which included three patients and
six subsequent pregnancies, reported an anamnestic response of the inhibitor in four of
the pregnancies. Therefore, it is considered essential to counsel women about this risk in
subsequent pregnancies (Delgado et al. 2003).
Autoimmune diseases
Inhibitors to factor VIII have been associated with the following derangements of the
immune system:
•
rheumatoid arthritis
•
systemic Sjögren’s syndrome
•
dermatomyositis
•
graft-versus-host disease after allogenic bone marrow transplantation
•
myasthenia gravis
•
multiple sclerosis
•
Grave’s disease
98
Use of recombinant and plasma-derived Factor VIII and IX
•
autoimmune haemolytic anaemia.
Patients with autoimmune disease-related inhibitors usually have high titre inhibitors that
require immediate treatment, as they do not spontaneously disappear. Prednisolone, a
synthetic corticosteroid with potent anti-inflammatory activity, shows a greater effect
with the addition of an alkylating agent that has immunosuppressant action (Delgado et
al. 2003).
Malignant neoplasms
There are inconsistencies between studies on whether the association between malignant
neoplasms (either solid or haematologic) and acquired haemophilia is causal, or whether
the appearance of solid neoplasms is simply common in elderly people in whom
inhibitors usually appear. After a review of case reports of 41 cancer-associated
inhibitors, the following recommendations were made: The primary malignancy should
be treated, as the antibodies are easier to eradicate when the tumour is controlled. If the
tumour is still present or has failed to respond to treatment, immunosuppressive therapy
should be considered. The decision to administer immunosuppressive therapy should
take into account the age, type of malignancy and severity of bleeding (Level IV
evidence) (Delgado et al. 2003).
Drug administration
The association between acquired haemophilia and drug administration is often unclear,
particularly in elderly people who are on multiple medications, including antibiotics and
anticonvulsants that have well-established associations with inhibitors. In most cases, the
inhibitor disappears when the drug is withdrawn (Delgado et al. 2003).
Previous Australian consensus-based guidelines
Currently there are no Australian guidelines on treatment of acquired haemophilia.
Effectiveness and safety of treatment of bleeding episodes
The treatment of acquired haemophilia is similar to that used for congenital haemophilia
with inhibitors. There are two fundamental issues: 1) immediate treatment of acute
bleeding episodes, as they can be life threatening; and 2) ultimate treatment to remove
inhibitors and thus, eradicate the disease (Stasi et al. 2004).
Current options for treating bleeding episodes in patients with inhibitors include the use
of high doses of factor VIII (plasma-derived or recombinant) or DDAVP if the inhibitor
levels are low (<5 BU). If the inhibitor levels are high (>5 BU) bypassing agents such as
aPCCs or rFVIIa should be used. See Table 34 for full details of products used in
patients with inhibitors.
Desmopressin (1-deamino-D-arginine vasopressin, DDAVP)
In a systematic review of low level evidence, desmopressin was shown to be effective in
patients with low titre inhibitors (<3 BU; Level IV evidence). However, a trial of
desmopressin in patients with high titre inhibitors or severe bleeding only delayed the
commencement of more effective treatments (Delgado et al. 2003).
Use of recombinant and plasma-derived Factor VIII and IX
99
Human FVIII concentrates
Human factor VIII (recombinant or plasma derived; rFVIII or pdFVIII) may be useful
in large doses for patients with low inhibitor titres (<5 BU), in a recommended dose of
20 IU/kg for each BU of inhibitor plus an additional 40 IU. If the initial dose is not
adequate after 10–15 minutes, another bolus injection may be administered. Some
clinicians used triple the dose of regular treatment with factor concentrates for
haemophilia and found clinical benefit despite poor post-infusion assay results. Response
to human factor VIII is highly variable in the presence of inhibitors and should be
closely monitored (Delgado et al. 2003). This finding is consistent with that reported in a
systematic review on the treatment of haemophilia A patients with inhibitors (Lloyd
Jones et al. 2003).
Activated prothrombin complex concentrates (aPCCs)
A recent retrospective case series (Level IV evidence) of 34 acquired haemophilia
patients found that a dose of 75 IU/kg of FEIBA every 8–12 hours was successful in
curbing bleeding episodes in 76% of severe and 100% of moderate episodes (Sallah
2004). The recommended dose for both FEIBA and Autoplex® is 50–200 IU/kg/day in
divided doses. To date, there is no available assay to measure the efficacy of aPCCs
(Delgado et al. 2003).
Overall, aPCCs appear to be well tolerated with very few side effects. However, patients
should be closely monitored as larger doses may cause thrombotic events, including
myocardial infarction, disseminated intravascular coagulation and thrombosis (Delgado
et al. 2003).
Activated recombinant FVII (rFVIIa)
The largest case series (Level IV evidence) that investigated the use of rFVIIa in patients
with acquired haemophilia reported that it was effective as a ‘good’ salvage therapy in
75% or ‘partial’ salvage therapy in 17% of bleeding episodes once other therapies had
failed. When used as first-line therapy, there was a 100% success rate. The average dose
of rFVIIa was 90 µg/kg every 2–6 hours for a median of 3.9 days (Delgado et al. 2003).
From a study of 180 000 standard doses of rFVIIa, it was concluded that rFVIIa is very
well tolerated, with few side effects. Close monitoring of patients is required as adverse
events (though rare) may be serious.
Eradication therapy
Immunosuppressants
In approximately 36% of acquired haemophilia cases, inhibitors disappear spontaneously
without immunosuppressive therapy (Delgado et al. 2003). However, complete remission
is unpredictable and it is recommended that patients be treated with immunosuppressive
therapy to assist in removal of inhibitors. However, specific recommendations cannot be
made about the safest and most effective type of immunosuppressive therapy for
acquired haemophilia due to the lack of quality evidence (Table 36).
Prednisolone in a dose of 1 mg/kg/day has been shown to abolish inhibitors in
approximately 30% of patients (Delgado et al. 2003). With the addition of
100
Use of recombinant and plasma-derived Factor VIII and IX
cyclophosphamide (1–2 mg/kg/day) the success rate increases to 60–100%.
Combinations of prednisolone with other immunosuppressants, such as azathioprine,
cyclophosphamide, or vincristine have also been effective. Cyclosporin A (200–300
mg/day) was found to be successful as a salvage therapy, alone or with prednisolone.
More recently, rituximab (375 mg/m2/week) has been reported to show effectiveness
after other treatments failed. All patients with inhibitor levels <100 BU achieved
complete remission, while the two patients with inhibitors >100 BU achieved partial
remission (Level IV evidence) (Stasi et al. 2004).
Immunosuppressive therapy may have multiple side effects in an elderly population (the
majority of acquired haemophilia cases). In a consecutive series of 18 patients with
acquired haemophilia (Level IV evidence), it was reported that one patient died from
bleeding, while three patients died from complications associated with
immunosuppressive treatment (Collins et al. 2004). Cyclophosphamide may cause
bacterial sepsis from neutropaenia, fibrotic lung disease, and a combination of
cyclophosphamide, prednisolone and vincristine has been reported to cause alopecia.
Prednisolone and cyclophosphamide have been associated with the development of
herpes zoster, cataracts, myelodysplasia and severe osteoarticular infections. A metaanalysis of case series of 249 patients found that, while cyclophosphamide was superior
to prednisolone for eliminating inhibitors, it was inferior in terms of survival. That is, a
substantial number of patients died from neutropaenia-related infections as a side effect
of cyclophosphamide treatment (Delgado et al. 2003). A more recent study (Level IV
evidence) on rituximab found it was associated with fewer adverse events than
cyclophosphamide, but these treatments have not been compared in a systematic way. A
total of 15 patients with acquired haemophilia have been treated with rituximab, with
very few side effects (Level IV evidence) (Stasi et al. 2004; Wiestner et al. 2002). Early
treatment and a combination of rituximab and prednisolone achieved optimal benefit
(Level IV evidence) (Wiestner et al. 2002).
Use of recombinant and plasma-derived Factor VIII and IX
101
Table 36.
Adverse events associated with immunosuppressant therapy
Study
Level and quality of
evidence
Population
Adverse events after immunosuppression
(Collins et al. 2004)
Level IV: case series
18 acquired
haemophilia: 14 no
underlying disorder,
1 gastric carcinoma,
1 polymyalgia
rheumatica, 1
Castlemanns
disease, 1 Depoxil;
aged 38–87, median
70 years
4 patients died: 1 from intracranial haemorrhage, and
3 from immunosuppressive-related treatment
6 acquired
haemophilia: 2 no
underlying disorder,
1 chronic
lymphocyctic
leukaemia, 1
scleroderma, 1
systemic lupus
erthematosus, 1
colon cancer; aged
59–78 years
2-chlorodeoxyadenosine as salvage therapy
Quality score: 3/3
Measurement bias
unlikely
Selection bias
avoided
Follow-up adequate
Uncontrolled
(Sallah & Wan 2003)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Follow-up adequate
Uncontrolled
(Delgado et al.
2002)
Level IV: case series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Follow-up adequate
Uncontrolled
17 acquired
haemophilia: 7 no
underlying disorder,
4 postpartum, 1
acute hepatitis B, 1
lymphoma, 1 lung
cancer, 1 refractory
anaemia with ringed
sideroblasts, 1
rheumatoid arthritis,
1 colon cancer; aged
8–86 years, median
67 years
Treatment was well tolerated, no grade 4 toxicity or
death from treatment observed:
1 case of mild neutropaenia
1 case with atypical pneumonia and lymphopaenia
9/17 (52%) had immunosuppressive therapy-related
side-effects:
3 died of immunosuppressive related side effects—2
with pneumonia, 1 with unspecified bacterial sepsis
Cyclophosphamide
Cytopaenia, hair loss, toxic hepatitis
Alkylating agent
Two episodes pancytopaenia, one cutaneous
reaction, one toxic hepatitis
Prednisolone
Steroid myopathy
Prednisolone + cyclophosphamide
3/7 had side effects, femoral nerve palsy,
neutropaenia, varicella zoster
2 deaths from pneumonia/septicaemia and
pulmonary fibrosis
(cont.)
102
Use of recombinant and plasma-derived Factor VIII and IX
Table 36 (cont.)
Level IV: case series
(Stasi et al. 2004)
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Wiestner et al.
2002)
Level IV: case series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Follow-up adequate
Uncontrolled
10 acquired
haemophilia: 6 with
no underlying
disorder, 1 with
rheumatoid arthritis,
1 with prostate
carcinoma, 1 with
low-grade nonHodgkin lymphoma,
and one after
pregnancy; aged 27–
78 years, median 61
years
4 acquired
haemophilia: 1
chronic renal failure,
1 ascites and
lymphadenopathy, 1
with polymyalgia and
ecchymoses, and 1
with no underlying
disorder. Aged 38–
70 years, median 54
years
Rituximab
3/10 patients had first-infusion reactions of fever (1
patient), chills (2 patients), and nausea (1 patient).
Reactions were brief and mild.
Rituximab
No side effects
High-dose intravenous immunoglobulin G
The literature on the effectiveness of intravenous immunoglobulin G (IVIg) in patients
with acquired haemophilia reported an overall patient response rate of 12–50%. The
current clinical evidence does not support the use of IVIg as first-line therapy (Level IV
evidence). Reported positive effects following the addition of IVIg to steroids were
probably due to publication bias (expert opinion) (Delgado et al. 2003).
Plasmapheresis and immunoadsorption
Plasmapheresis and immunoadsorption (extracorporeal removal of antibodies) may be an
option when severe bleeding results in the need for rapid removal of inhibitors. With few
exceptions, these methods have been evaluated in conjunction with immunosuppression
therapy and their specific contribution to haemostasis is unclear. These methods have
not been found to reduce time to complete remission and should only be used with
immunosuppressive therapy to prevent the recurrence of inhibitors (expert opinion)
(Delgado et al. 2003).
Immune tolerance induction
While immune tolerance induction may be effective in patients with congenital
haemophilia and factor VIII alloantibodies, tolerisation is rarely if ever used in patients
with acquired haemophilia. Some studies have combined the use of factor VIII
concentrates and immunosuppressive agents to reduce inhibitors, but others found that
the immunosuppressive agents alone were as successful as when used in combination
with factor concentrates (Level IV evidence) (Delgado et al. 2003).
Use of recombinant and plasma-derived Factor VIII and IX
103
Evidence-based clinical practice guidelines
Treatment of bleeding episodes (All Level IV evidence)
•
It is recommended that bleeding episodes in patients with acquired haemophilia receive prompt
treatment.
When inhibitors <5 BU, DDAVP or FVIII concentrate should be used:
− DDAVP may be used in a dose 0.3 µg/kg body weight, either intravenously or subcutaneously. It
should be diluted in 50 mL of 0.9% saline and infused over at least 30 minutes.
− FVIII concentrate may be used in a recommended dose of 20 IU/kg for each BU of inhibitor plus
an additional 40 IU and tested after 15 minutes. If response is poor, another bolus injection may
be given.
− Activated recombinant factor VII (rFVIIa) may be given in a dose of 90 µg/kg every 2–6 hours
until bleeding has stopped.
When inhibitors >5 BU, activated prothrombin complex concentrates (aPCCs) or rFVIIa should be
used:
− aPCCs may be given in a dose of 50–200 IU/kg/day in divided doses.
− rFVIIa may be given in a dose of 90 µg/kg every 2–6 hours until bleeding has stopped.
•
Regardless of treatment option taken, it is recommended that the patient be closely monitored due
to the risk of adverse thrombotic events.
Elimination of inhibitors (All Level IV evidence)
•
It is recommended that patients with acquired haemophilia be examined for underlying disorders
before deciding on treatment strategies. The decision to administer immunosuppressive therapy
should take into account the age, type of malignancy and severity of bleeding.
− Patients with postpartum acquired haemophilia (without lupus or rheumatoid arthritis) may
receive immunosuppressive therapy, or may simply be monitored, due the high rate of
spontaneous disappearance of inhibitors within this group.
− Patients with drug-related acquired haemophilia should have the drug withdrawn, in the first
instance, and be treated with immunosuppressive therapy or simply monitored for spontaneous
disappearance of inhibitor.
− Patients with autoimmune disease-related inhibitors usually have high titre inhibitors that require
treatment with immunosuppressants.
− Patients with acquired haemophilia associated with a malignant neoplasm should have the
primary malignancy treated initially, as the antibodies are easier to eradicate once the tumour is
controlled. Immunosuppressive therapy should be considered while the tumour is still present or
has failed to respond to treatment.
− Patients without underlying disorders should receive immunosuppressive therapy.
•
104
Due to the lack of quality evidence on effectiveness and safety of particular immunosuppressants,
no recommendation can be made regarding the best treatment, but patients should be treated on
an individual basis with close monitoring for adverse events.
Use of recombinant and plasma-derived Factor VIII and IX
Surgical and Dental Procedures
Haemophilia A and B
Surgical procedures
Before the 1960s, the mortality rates for haemophilia patients undergoing surgery were
very high (25–50%) (Bastounis et al. 2000). Mortality decreased with the use of plasma
(to <10%), cryoprecipitate (to <2%), and the more recent introduction of factor
concentrates (to <1%).
Major surgical procedures in patients with coagulation disorders, particularly in those
with inhibitors, carry substantial risks and should be undertaken in a haemophilia care
centre with careful individual management after consultation with a haemophilia
specialist.
There were no studies available that formally looked at the optimal duration of treatment
after surgical procedures.
Previous Australian consensus-based guidelines
There are no existing Australian guidelines for the management of patients undergoing surgical
procedures. Recommendations from the Canadian guidelines are listed in Table 37.
Table 37.
Management of patients undergoing surgical procedures
Before surgery
•
If the risk of bleeding is very high, elective surgery should be postponed
•
Screening for inhibitors should be performed shortly before the day of surgery (Bethesda assay)
•
The patient should avoid using antiplatelet medication before and after surgery
•
Surgery should be scheduled for early in the week and early in the day to ensure availability of laboratory services and
consultants
•
Adequate amounts of replacement factor product required before and after surgery should be available
•
A pharmacokinetic analysis to determine recovery/response to a challenge dose of factor product should be conducted
before surgery
During surgery
•
Caution should be taken to avoid medications or procedures that provoke haemorrhages (e.g. analgesics, anaesthetics;
ligation of all visible bleeding, cautery only for capillary bleeding)
•
Keep operation time to a minimum
•
Use pressure bandages for soft tissue injuries
After surgery
•
Careful monitoring (factor activity levels) to maintain plasma factor levels >30% for the period of recovery after surgery (2–
3 weeks)
•
Remove sutures after factor replacement given
Modified from Association of Hemophilia Clinic Directors of Canada 1995a; Bastounis et al. 2000. These guidelines do not match with current
Australian practice (AHCDO Executive Committee 2005)
Use of recombinant and plasma-derived Factor VIII and IX
105
Are treatments effective?
Comparisons between patients and across studies are difficult because of considerable
variability in surgical procedures (minor/major; elective/emergency), duration of therapy,
dosing requirements (pre-operative/additional doses, bolus/continuous dosing).
All available studies (Table 38) relevant to the use of rFVIII or rFIX in patients
undergoing surgery were low level evidence (Level IV). Haemostatic control was
described by physicians as ‘good’ or ‘excellent’ in all patients, without need for additional
treatment. Overall, blood loss was estimated to be within the normal range expected in
patients without a coagulation disorder for the type of surgery performed.
106
Use of recombinant and plasma-derived Factor VIII and IX
Table 38.
Haemostatic control in haemophilia patients undergoing surgery
Study
Level and quality of
evidence
Population
(Scharrer 2002;
Scharrer et al. 2000)
Level IV: before-andafter study
15 PTPs and 7 PUPs
with severe
haemophilia A,
without inhibitors,
undergoing 30
surgical procedures
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Schwartz et al.
1990; Seremetis et
al. 1999)
Level IV: case series
Quality score: 2/3
Selection bias unclear
Confounding avoided
Follow-up adequate
Uncontrolled
26 PTPs with
moderate–severe
haemophilia A,
without inhibitors,
undergoing 32
surgical procedures,
or with serious
haemorrhages
Haemostatic control
PTPs
PUPs
Haemostasis
control
18/23 excellent
5/23 good
7/7 excellent
Median postsurgical
duration of
therapy
12 days (1–89)
8 days (5–12)
Median
number of
infusions
18 (1–75)
12 (6–44)
Daily dose
52.8 IU/kg/day
(10.6–149.4
IU/kg/day)
93 IU/kg/day
(51–235
IU/kg/day)
Blood loss
0–250ml within
normal range
expected in
patients without
coagulation
disorder
within normal
range
100% rated ‘excellent’—no additional treatment
required
rFVIII treatment as
required
(Ragni et al. 2002)
Level IV: before-andafter study
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
26 PTPs with mild–
severe haemophilia B
and 2 female
haemophilia B
carriers, undergoing
32 surgical
procedures
Intra-operative blood loss:
13 PTPs with
moderate-severe
haemophilia A,
without inhibitors
undergoing surgical
procedures
Blood loss: 0–200ml, as expected for the type of
surgery in patients without coagulation disorder
16 PTPs with mildsevere haemophilia
A, without inhibitors,
undergoing 20
surgical procedures
Haemostasis control:
Post-operative blood loss:
Haemostatic control:
0–1500ml
0–2676ml
excellent/good 34/35
Uncontrolled
(White et al. 1997)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Confounding avoided
Follow-up adequate
Uncontrolled
(Stieltjes et al. 2004)
Level IV:
retrospective case
series
Quality score: 1/3
Selection bias
uncertain
Follow-up adequate
excellent 11/20
good 5/20
moderate 4/20
Blood loss:
<500ml 6/20
>500ml 3/20
Excessive bleeding required red blood cell
transfusion in 8 procedures
Uncontrolled
PTPs = previously treated patients; PUPs = previously untreated patients; rFVIII= recombinant factor FVIII
Use of recombinant and plasma-derived Factor VIII and IX
107
Dental surgery
Dental procedures for patients with bleeding disorders require consultation between the
patient’s dentist and a haemophilia specialist.
Previous Australian consensus-based guidelines
Protocols for the dental management of coagulation disorders, haemophilia A, B and von
Willebrand disease have been developed and implemented at the Adelaide Dental Hospital in
South Australia (Stubbs & Lloyd 2001) (Appendix G).
Are treatments effective?
Data on treatment effectiveness is limited. Over a two year period, 30 haemophilia A,
one haemophilia B, and 15 von Willebrand patients were managed successfully according
to the protocol devised at the Adelaide Dental Hospital in South Australia (no data
provided) (Stubbs & Lloyd 2001). The extent of treatment for controlling haemostasis
was based on the expected degree of trauma to oral soft tissue during dental procedures
and the severity of disease in the patient. The management of dental extractions was not
included in the protocol.
One average quality randomised controlled trial (Level II evidence) investigated the
effect of suturing the tooth extraction wound compared to leaving it open in two groups
of patients with haemophilia A (Stajcic et al. 1989). Although there was no significant
difference between groups in the incidence of post-operative bleeding, the clot size,
which was measured subjectively (unvalidated measure), was significantly larger in the
group with open wounds (p<0.01), making the post-operative period more
uncomfortable for patients.
Table 39.
Haemostatic control in haemophilia patients undergoing dental procedures
Study
Level and quality of
evidence
(Stajcic et al. 1989)
Level II: randomised
controlled trial
Quality score: 14/27
Selection bias
minimised
Confounding
uncertain
Follow-up adequate
Population
Haemostatic control
62 patients with mildsevere haemophilia
A, undergoing dental
extractions
Open wound
(N=25)
Closed
wound
(N=25)
Clot size large
15/25
1/22
Post-operative
bleeding
2/25
0/25
Post-operative
dose FVIII
2.7 IU/kg
0
Patients
representative
Are treatments safe for surgery or dental surgery?
Inhibitor development following treatment with rFVIII or rFIX was either low titre and
transient or not detected in two low level studies described in Table 40.
108
Use of recombinant and plasma-derived Factor VIII and IX
Table 40.
Development of inhibitors in haemophilia patients undergoing surgical procedures
Study
Level and quality of
evidence
Population
(Ragni et al. 2002)
Level IV: case series
26 PTPs with mild–
severe haemophilia B
and 2 female
haemophilia B
carriers, undergoing
32 surgical
procedures
None detected
15 PTPs and 7 PUPs
with severe
haemophilia A,
without inhibitors,
undergoing surgical
procedures
High-titre inhibitor
Low-titre inhibitor
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
Uncontrolled
(Scharrer 2002;
Scharrer et al. 2000)
Level IV: case series
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
Development of inhibitors associated with
treatment
0
2/7 PUPs
2 transient (prior to
surgery)
Uncontrolled
PTPs = previously treated patients; PUPs = previously untreated patients
Overall, there were few adverse events following surgery (Table 41). Although most
adverse events were mild and reversible, Stieltjes et al. 2004 reported four serious postoperative bleeding complications following major surgery (Level IV evidence).
Some dental patients treated according to the protocol developed by Stubbs and Lloyd
(2001) reported nausea following the use of tranexamic acid. Generally, there were no
post-operative bleeding complications following dental treatment. In the majority of
patients, gingival ooze stopped within 60 minutes; some patients with severe haemophilia
A experienced mild gingival ooze for several hours post-operatively.
Use of recombinant and plasma-derived Factor VIII and IX
109
Table 41.
Adverse events in haemophilia patients undergoing surgical or dental procedures
Study
Level and quality of
evidence
(Zanon et al. 2000)
Level III-2: cohort
study
Quality score: 16/27
Selection bias
uncertain
Population
71 haemophilia A and
6 haemophilia B
patients undergoing
dental extractions
Controls: 184 nonhaemophilic patients
undergoing dental
extractions
Confounding
uncertain
Adverse events
No. of bleeding
complications/No of
dental extractions
Haemophilia patients
(N=75)
2/98 (2.0%)
Non-haemophilia
controls (N=184)
1/239 (0.4%)
Follow-up adequate
Patients
representative
(Ragni et al. 2002)
Level IV: before-andafter study
Quality score: 2/3
Selection bias
minimised
Follow-up adequate
26 PTPs with mild–
severe haemophilia B
and 2 female
haemophilia B
carriers, undergoing
32 surgical
procedures
4 patients experienced ≥1 adverse event
Rash, flushing, hives, headache, teichopsia,
sneezing, dry cough, local phlebitis
Uncontrolled
(Scharrer 2002;
Scharrer et al. 2000)
Level IV: before-andafter study
Quality score: 2/3
Selection bias
minimised
15 PTPs and 7 PUPs
with severe
haemophilia A,
without inhibitors,
undergoing surgical
procedures
No adverse events or complications
8 patients with
haemophilia B
undergoing dental
extraction
4/8 patients reported adverse effects of EACA
treatment:
16 PTPs with mild–
severe haemophilia
A, without inhibitors,
undergoing 20
surgical procedures
5 mild–moderate and 4 serious adverse events
associated with BDDrFVIII treatment
No adjunctive agents required
Follow-up adequate
Uncontrolled
(Djulbegovic et al.
1996)
Level IV: case series
Quality score: 2/3
Selection bias
uncertain
Nausea, vomiting, transient creatine
phosphokinase elevation
Follow-up adequate
Uncontrolled
(Stieltjes et al. 2004)
Level IV:
retrospective case
series
Quality score: 1/3
Selection bias
uncertain
Mild–moderate: haematoma, obturation of the vein
at infusion site, local thrombophlebitis at infusion
site
Serious: severe postoperative bleeding
Follow-up adequate
Uncontrolled
BDDrFVIII = recombinant B-domain deleted FVIII; EACA = ε-aminocaproic acid, an antifibrinolytic agent; PTPs = previously treated patients;
PUPs = previously untreated patients
von Willebrand disease
One of the most common symptoms of von Willebrand disease is excessive postoperative bleeding. Therefore, dental and surgical procedures should be performed with
caution in this population (Ziv & Ragni 2004). Bleeding episodes in people with von
Willebrand disease (vWD) may be treated either with DDAVP or factor VIII
concentrates enriched with von Willebrand factor (pdFVIII/vWF). Optimal treatment
110
Use of recombinant and plasma-derived Factor VIII and IX
depends on the type of vWD and the patient’s responsiveness to DDAVP (see
Adjunctive haemostatic agents section).
Are treatments effective?
Local therapies and DDAVP
There were seven low level research papers included in this systematic review that
evaluated the effectiveness of the use of DDAVP plus local therapies for dental or
surgical procedures (See Table 42 and Table 43). Overall, it was found that a
combination of DDAVP and local therapiesx such as tranexamic acid and fibrin glue was
highly effective for minimising post-operative bleeding (Level IV evidence) (Mariana et
al. 1984; Ghirardini et al. 1988; de la Fuente et al. 1985; Warrier & Lusher 1983; NituWhalley et al. 2001; Saulnier et al. 1994).
Table 42.
Study
(Saulnier et al.
1994)
Effectiveness of adjunctive haemostatic agents in surgical and dental procedures for
patients with von Willebrand disease
Level and quality
of evidence
Effectiveness of DDAVP + local therapies
Level IV: before and
after study
14 vWD, mean age 24
years
DDAVP (0.3 µg/kg) + tranexamic acid + fibrin glue
Quality score: 2/3
16 treatments, 14 dental
extractions
Number without bleeding complications:
100%
FVIII:C mean increase post/pre treatment
X4.6
Selection bias
minimised
vWF:Ag mean increase post/pre
treatment
X3.22
Follow-up adequate
RcoF mean increase post/pre treatment
X4.08
Uncontrolled
BT mean decrease (measured by Duke
method) post/pre treatment
0.27
Measurement bias
possible
(Nitu-Whalley et
al. 2001)
Population
27 patients with Types 1
(93%) and 2 (7%) vWD
most of whom had
responded to DDAVP,
aged 14–57 years
Intravenous DDAVP (0.3 µg/kg) + tranexamic acid
for mucosal surgery
35 surgical events,
including major, minor
and dental
‘Moderate’ after 2/35, and
Level IV: case
series
11 vWD patients:
DDAVP (0.3 µg/kg) intravenous + epsilon
aminocaproic acid
Quality score: 2/3
3 x Type 2A vWD
Level IV: case
series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Haemostasis rated as
‘Excellent’ after 32/35 surgical events,
‘Poor’ after 1/35 surgical event.
Follow-up adequate
Uncontrolled
(de la Fuente et
al. 1985)
Measurement bias
minimised
Selection bias
possible
8 x Type 1 vWD,
6 dental procedures, 9
surgical interventions
Follow-up adequate
Excellent haemostasis achieved in all 5 patients who
had dental extractions
4/6 patients who had surgical procedures had no
need for factor concentrates
2/6 patients required fresh frozen plasma or
cryoprecipitate due to low FVIII:C levels after surgery.
Uncontrolled
x
(cont.)
Local therapies—application of a treatment to the bleeding site
Use of recombinant and plasma-derived Factor VIII and IX
111
Table 42 (cont.)
Study
(Warrier &
Lusher 1983)
Level and quality
of evidence
Level IV: case
series
Quality score: 2/3
Measurement bias
minimised
Selection bias
possible
Population
7 patients with vWD
7 surgical events,
including 3 dental
extractions, 3
tonsillectomies, 1 nasal
polypectomy
Follow-up adequate
Effectiveness of DDAVP + local therapies
DDAVP via intranasal drip (2–4 µg/kg) or
intravenous (0.2–0.3 µg/kg) + epsilon
aminocaproic acid
No unusual bleeding from any of the 7 surgical events
6/15 had prolonged BT at baseline
All 6 had normal values 15 and 90 minutes post
DDAVP
5/6 had prolonged BT at 180 minutes
Uncontrolled
(Ghirardini et al.
1988)
Level IV: case
series
Quality score: 2/3
5 Type 1 vWD patients
1 dental extraction, 4
surgical interventions
Measurement bias
minimised
DDAVP subcutaneous (0.3 µg/kg) + tranexamic
acid + fibrin glue for dental
No bleeding observed in any dental or surgical
procedure
Selection bias
possible
Follow-up adequate
Uncontrolled
(Mariana et al.
1984)
Level IV: case
series
Quality score: 1/3
Measurement bias
possible
20 vWD (Types 1 and 2)
9 patients had 12 dental
extractions, 6 patients
had 8 surgical
procedures
Selection bias likely
Follow-up adequate
DDAVP (0.3–0.4 µg/kg) intravenous + tranexamic
acid
No bleeding observed in vWD patients for dental
procedures
No intra-operative bleeding in 5/6 patients post
surgery. One patient had a bleeding episode on the
7th day after surgery
Uncontrolled
BT= bleeding time; DDAVP= 1-D-deamino-8D-arginine vasopressin; FVIII:C= factor VIII coagulant activity; RcoF = ristocetin-cofactor activity;
vWF:Ag= von Willebrand factor antigen; vWD= von Willebrand disease
Table 43.
Effectiveness of local therapies, local therapies with DDAVP, and local therapies with factor
concentrates for dental surgery in von Willebrand disease patients
Study
Level and quality of
evidence
(Federici et al.
2000)
Level IV: retrospective
case series
Quality score: 2/3
Measurement bias
possible
Selection bias
minimised
Population
63 patients with vWD,
for 117 dental
extractions or
periodontal surgery
episodes
31 x Type 1
22 x Type 2
10 x Type 3
Effectiveness of tranexamic acid ± fibrin glue +
DDAVP or factor VIII/vWF concentrates
Local
therapies
alone
Local
therapies +
DDAVP
Local
therapies +
pdFVIII/vWF
1/30 patients
experienced
bleeding
complications
0/66 patients
experienced
bleeding
complications
1/21 patients
experienced
bleeding
complications
Follow-up adequate
Uncontrolled
DDAVP= 1-D-deamino-8D-arginine vasopressin; vWD= von Willebrand disease; vWF = von Willebrand factor
Plasma-derived factor VIII/von Willebrand factor concentrates
Seven case series (Level IV evidence) evaluated the effectiveness of using pdFVIII/vWF
concentrates for haemostasis cover during surgical or dental procedures in patients with
von Willebrand disease. The studies varied in amount of factor product used prior to and
112
Use of recombinant and plasma-derived Factor VIII and IX
after surgery but, overall, pdFVIII/vWF concentrates were found to provide
excellent/good haemostatic cover for surgical or dental procedures (Table 44).
One low level study reported that all of the 73 surgical cases received Haemate® P in a
single dose prior to surgery and, by day seven, only 11/73 (15%) of cases required
further treatment (Lillicrap et al. 2002; Dobrkovska et al. 1998). An ‘excellent/good’
physician rating was achieved in 99% of cases, indicating that haemostasis and adequate
control of bleeding had been achieved. Individual assessment and monitoring of the
patient was found to be essential during surgical procedures, due to the wide range of
doses required for haemostasis (Level IV evidence) (Lillicrap et al. 2002; Dobrkovska et
al. 1998).
As reported in Thompson et al. 2004, a pre-operative loading dose of 60–80 IU
vWF:Rco/kg (27-36 IU FVIII:C/kg) was administered every 8–12 hours for three days
(Level IV evidence). Maintenance doses were usually administered for seven days postsurgery but treatment was extended in 10/32 (31.3%) patients for more than seven days.
Patients treated for longer periods tended to be given lower, less frequent doses. The
wide range in dose, number of infusions and duration of treatment indicate that
treatment must be assessed on an individual basis, and levels of von Willebrand factor
activity and factor VIII need to be measured to tailor individual treatment programs. No
consistent pattern was observed for loading or maintenance doses by von Willebrand
disease type. However, Type 3 patients appeared to receive a greater number of infusions
(13). In addition, the majority of patients with Type 1 and 2 von Willebrand disease
completed treatment within seven days (Thompson et al. 2004).
Table 44.
Effectiveness of plasma-derived factor VIII/von Willebrand factor concentrates for surgery or
dental procedures in von Willebrand patients
Study
Level and quality
of evidence
(Auerswald et al.
2002)
Level IV: beforeand-after study
Quality score: 3/3
Measurement
bias unlikely
Population
Effectiveness
14 patients with
vWD, treated with
Immunate® for 14
acute bleeds or
surgical events
Haemostasis ‘Excellent’ or ‘Good’ in all patients after treatment
39 patients with
vWD, treated with
Alphanate® for
71 surgical or
invasive
procedures
Antifibrinolytic required in 8/71 (11.3%) procedures, mainly in oral
cavity
Normal perioperative bleeding in most cases
3/14 patients experienced post operative complications, 2 related to
insufficient cover and were resolved by further factor concentrate
administration
Selection bias
minimised
Follow-up
adequate
Uncontrolled
(Mannucci et al.
2002)
Level IV:
prospective case
series
Quality score: 3/3
Measurement
bias unlikely
Selection bias
minimised
Follow-up
adequate
6 x Type 1
17 x Type 2A
2 x Type 2B
14 x Type 3
Median number of infusions= 3
Median treatment interval across all infusions= 24 hours
Prolonged bleeding time corrected 30 minutes post infusion in 25/63
patients (39.7%) and partially corrected in 25/63 patients (39.7%),
no correction in 12/63 (19/0%)
Uncontrolled
Use of recombinant and plasma-derived Factor VIII and IX
(cont.)
113
Table 44 (cont.)
Study
Level and quality
of evidence
Population
(Gill et al. 2003)
Level IV:
prospective case
series
33 patients with
vWD, treated with
290 infusions of
Haemate® P for
53 bleeding
episodes—both
surgical and
spontaneous
bleeding episodes
Quality score: 3/3
Measurement
bias unlikely
Selection bias
minimised
Follow-up
adequate
Uncontrolled
(Lillicrap et al.
2002; Dobrkovska
et al. 1998)
52/53 (98%) bleeding episodes had ‘excellent/good’ haemostasis
after treatment
9 x Type 1
4 x Type 2A
7 x Type 2B
27 x Type 3
6 x Other
Level IV:
retrospective case
series
97 patients with
vWD treated for
73 surgical events
Quality score: 2/3
32 x Type 1
Measurement
bias possible
5 x Type 2A
Selection bias
minimised
28 x Type 3
Follow-up
adequate
Effectiveness
72/73 (98.6%) surgical cases had an ‘excellent/good’ outcome
18 x Type 2B
14 x Other
Uncontrolled
(Thompson et al.
2004)
Level IV:
prospective case
series
39 patients with
vWD with 42
surgical events:
Overall physician-rated effectiveness was ‘excellent/good’ for 39/39
(100%)
Quality score: 2/3
17 minor and 25
major surgeries
Number of infusions
Measurement
bias minimised
Selection bias
possible
Follow up
adequate
Uncontrolled
16 x Type 1
4 x Type 2A
5 x Type 2B
8 x Type 3
6 x Other
Daily physician ratings were ‘excellent/good’ for 38/39 (97.4%)
39 patients required 354 infusions of Haemate® P
Median average daily maintenance dose per infusion = 52.8 IU kg-1
(range 24.2–196.5 IU kg-1)
Median number of infusions per surgical event = 6 (range 1–67
infusions)
Median number of days with treatment = 3 days (range 1–50 days)
Median treatment duration = 3 (range 1–75 days)
Number of Infusions by von Willebrand Type
Mean per patient
(range)
Mean per day per
patient
Type 1 (n=17)
3 (1–20)
0.97
Type 2A (n=4)
4 (3–19)
0.86
Type 2B (n=5)
7 (2–9)
1.53
Type 3 (n=9)
13 (1–67)
1.18
Other (n=4)
8 (1–13)
1.0
(cont.)
114
Use of recombinant and plasma-derived Factor VIII and IX
Table 44 (cont.)
(Franchini et al.
2003)
Level IV:
retrospective case
series
Quality score: 1/3
Measurement
bias possible
Selection bias
possible
26 patients with
von Willebrand
disease, treated
for 43 surgical or
invasive
procedures
19 x Type 1
7 x Type 2B
vWF:Rco a
vWF:Ag a
FVIII:C a
Mean pre-infusion
IU/dL (range)
12.4 (0–36)
Mean pre-infusion
IU/dL (range)
17 (0–76)
Mean pre-infusion
IU/dL (range)
30 (6–78)
1-hour after preoperative infusion
IU/dL (range)
134.7 (78–265)
1-hour after preoperative infusion
IU/dL (range)
166.6 (90–299)
1-hour after preoperative infusion
IU/dL (range)
112.7 (62–226)
Mean IVR (range)
2.0 (1.4–2.7)
Follow-up
adequate
Amount of Haemate® P used
Uncontrolled
Mean (range)
(Federici et al.
2002)
Level IV:
retrospective case
series
Quality score: 1/3
Measurement
bias possible
13/22 patients
with von
Willebrand
disease, treated
with Fanhdi® for
14 invasive
procedures
Unable to assess
selection bias
9 x Type 1
Follow-up
adequate
6 x Type 3
Type of
procedure
Total dose (IU
vWF:Rco/kg)
Days of
treatment
Daily dose (IU
vWF:Rco/kg/day)
Major
surgery
284.1
(125.0–976.4)
9.7
(5–23)
39.3
(25.0–52.5)
Minor
surgery
120.8
(42.9–173.3)
4.2
(2.7)
28.7
(21.4–34.8)
Dental
extractions
38.4
(23.5–100.0)
1.6
(1–5)
24.0
(23.5–25.0)
Invasive
procedures
87.3
(27.3–160.0
2.7
(1–5)
32.3
(27.3–37.0)
Total
183.2
(23.5–976.4)
5.7
(1–23)
31.5
(21.4–52.5)
Type of procedure
Subjective clinician rated
haemostasis
Major surgery
‘Excellent/good’ in 7/7 procedures
Minor surgery
‘Excellent/good’ in 5/5 procedures
Dental extractions
‘Excellent/good’ in 1/2 procedures
One Type 3 vWD patient had poor
outcome
7 x Type 2B
Uncontrolled
results expressed as mean (range); FVIII:C = factor VIII; IVR = in vivo recovery (IU dl-1 per IU kg-1); RcoF = ristocetin-cofactor activity; vWD=
von Willebrand disease; vWF:Ag = von Willebrand factor antigen; vWF = von Willebrand factor
a
Are treatments safe?
DDAVP
The safety of DDAVP is discussed in an earlier section (See Adjunctive haemostatic
agents).
Plasma-derived factor concentrates
The safety of pdFVIII/vWF concentrates for von Willebrand disease was reported in
eight low level studies (Level IV evidence) that concluded that factors such as Haemate®
P, Immunate® and Fanhdi® (not currently available in Australia) are safe in preventing
excessive bleeding after major and minor surgery or invasive procedures (See Table 45).
However, due to the nature of plasma-derived concentrates, they still pose a theoretical
risk of viral and prion transmission.
Use of recombinant and plasma-derived Factor VIII and IX
115
Table 45
Adverse events associated with the use of plasma-derived factor VIII/vWF concentrates
Study
(Mannucci et al.
2002)
Level and quality of
evidence
Level IV: Prospective case
series
Quality score: 3/3
Measurement bias unlikely
Selection bias minimised
Follow-up adequate
Uncontrolled
(Gill et al. 2003)
81 vWD patients treated with
Alphanate®-Solvent
Detergent (A-SD) or
Alphanate®Solvent
Detergent/Heat-treated (ASD/HT) for 71
surgical/invasive procedures
or 87 bleeding episodes
Adverse events
A-SD
A-SD/HT
9/66 (13.6%) patients
had adverse events
5/36 (13.9%)
patients had
adverse events
Most adverse events were mild:
Reduced in vivo recovery, B19 parvovirus
transmission, thrombotic complications
15 x Type 1
29 x Type 2A
5 x Type 2B
32 x Type 3
Level IV: Prospective case
series
Quality score: 3/3
Measurement bias unlikely
Selection bias minimised
33 vWD patients, treated
with 290 infusions of
Haemate® P for 53 bleeding
episodes—both surgical and
spontaneous bleeding
episodes
24/53 bleeding or surgical events were
associated with adverse events
One mild allergic reaction was related to
treatment
9 x Type 1
4 x Type 2A
7 x Type 2B
27 x Type 3
6 x Other
Follow-up adequate
Uncontrolled
(Auerswald et al.
2002)
Population
Level IV: case series
14 vWD patients treated with
Immunate® for surgical or
bleeding episodes
No serious adverse events
39 vWD patients with 42
surgical treatment events
55 adverse events were reported in 24/42
(57.1%) of surgical treatments
16 x Type 1
4 x Type 2A
5 x Type 2B
8 x Type 3
6 x Other
4/55 (7.3%) included abdominal pain,
infection and abnormal wound healing not
related to treatment
Level IV: Prospective case
series
6 vWD patients treated with
Haemate® P
Quality score: 2/3
2 x Type 1B
2 x Type 2A
2 x Type 3
2/6 patients experienced mild adverse
events, including rash, hives, cough,
fever, weakness and nausea
Quality score: 3/3
Measurement bias unlikely
One phlebitic reaction at venous access
site after 102 hours of continuous infusion
Selection bias minimised
Follow-up adequate
Uncontrolled
(Thompson et al.
2004)
Level IV: case series
Quality score: 2/3
Measurement bias
minimised
Selection bias possible
Follow up adequate
Uncontrolled
(Dobrkovska et al.
1998)
Measurement bias unlikely
Selection bias possible
8/55 (14.5%) related to Haemate® P,
including paraesthesia, allergic reaction,
vasodilation, peripheral oedema,
extremity pain, pseudo-thrombocytopenia
and pruritus
Follow-up adequate
Uncontrolled
116
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 45 (cont.)
(Lillicrap et al.
2002; Dobrkovska
et al. 1998)
Level IV: Retrospective case
series
Quality score: 2/3
Measurement bias possible
Selection bias minimised
Follow-up adequate
Uncontrolled
(Franchini et al.
2003)
Level IV: Retrospective case
series
Quality score: 1/3
Measurement bias possible
Selection bias possible
97 vWD patients treated with
Haemate® P for 73 surgical
procedures, 344 bleeding
episodes, 20 prophylaxis
and 93 other events
16/97 (16.5%) patients experienced
adverse events within 24 hours of
Haemate® P administration
32 x Type 1
5x Type 2A
18 x Type 2B
28 x Type 3
14 x Other
26 vWD patients treated with
Haemate® P for 43 surgical
or invasive procedures
5/43 (11.6%) interventions (all major
surgery) required red blood cell
transfusion
19 x Type 1
7 x Type 2B
No adverse events associated with
Haemate® P treatment
13/22 vWD patients treated
with Fanhdi® for 14 invasive
procedures
No adverse events associated with
Fanhdi® treatment
Follow-up adequate
Uncontrolled
(Federici et al.
2002)
Level IV: Retrospective case
series
Quality score: 1/3
Measurement bias possible
Unable to assess selection
bias
9 x Type 1
7 x Type 2B
6 x Type 3
Follow-up adequate
Uncontrolled
A-SD = Alphanate® solvent/detergent treated; A-SD/HT = Alphanate® solvent/detergent, heat-treated; vWD=von Willebrand disease
Evidence-based clinical practice guidelines
On the basis of the available evidence, surgical and dental procedures in patients with haemophilia A, B
or von Willebrand disease should be managed as follows:
•
Recombinant FVIII is safe (Level III-2 evidence) and effective (Level IV evidence) for patients with
haemophilia A before and after surgery as required.
•
Recombinant FIX is safe and effective for patients with haemophilia B before and after surgery as
required (Level IV evidence).
•
Tranexamic acid may be used to control bleeding during surgical or dental procedures (Level IV
evidence).
•
DDAVP may be used alone or in conjunction with local therapies such as fibrin glue. A test dose of
DDAVP should be performed to determine responsiveness (Level IV evidence).
•
Plasma-derived FVIII/vWF concentrates may be used to control bleeding in patients with von
Willebrand disease who do not respond to DDAVP (Level IV evidence).
Use of recombinant and plasma-derived Factor VIII and IX
117
Delivery of Infants
Pregnancy, labour and delivery pose risks of bleeding complications to both the mother,
if she has von Willebrand disease or is a carrier of haemophilia A or B, and the newborn
infant with haemophilia. A variety of ethical and haemostatic challenges are presented
throughout the pregnancy, delivery and postpartum period, including invasive antenatal
diagnostic procedures, management of miscarriage, termination of pregnancy and the risk
of haemorrhage during labour and delivery. Carriers of haemophilia are defined as
obligate (father had haemophilia), presumed (more than one son has haemophilia),
potential (maternal relative has haemophilia), or sporadic (no family history—identified
through an affected child) (Kulkarni & Lusher 2001). Since congenital haemophilia is
linked to the X-chromosome, carriers usually have one normal gene that expresses
sufficient clotting factor to maintain normal haemostasis. However, in approximately 10–
20 per cent of carriers, extreme lyonisation may occur and very low levels of FVIII or
FIX (to <10% of normal) increase the risk of haemorrhage during delivery and/or
postpartum.
The main challenges are: 1) antenatal testing; 2) management of maternal coagulation
disorder during pregnancy; 3) management and method of delivery to prevent
haemorrhage in both mother and affected infant; and 4) control of haemorrhage (mother
and newborn) during postpartum period.
These guidelines assume a correct diagnosis and that the severity of the disorder has been
ascertained.
Previous Australian consensus-based guidelines
Currently, there are no Australian guidelines for the management of haemophilia carriers, pregnant
women with von Willebrand disease, or for the delivery of infants with haemophilia.
No available studies concerning the delivery of infants in other rare bleeding disorders,
such as fibrinogen abnormalities, or deficiencies in factors V, VII, X, XIII or
prothrombin, met the inclusion criteria for an evaluation of effectiveness or safety in this
review. In addition, no studies examining the safety of epidural analgesia (for the
haemophilia carrier or affected infant) during infant delivery met the inclusion criteria.
Effectiveness of antenatal diagnosis
The decision to have children is complex for carriers of the haemophilia gene due to the
well established genetic inheritance of haemophilia. Antenatal diagnosis can aid decision
making regarding pregnancy termination or guide the management of the pregnancy and
delivery. However, the uptake of antenatal diagnostic procedures in mothers with a
known risk is relatively low (approximately 35%) (Kadir et al. 1997).
There are several antenatal diagnostic procedures available at different stages of
pregnancy (Table 46).
118
Use of recombinant and plasma-derived Factor VIII and IX
Table 46.
Antenatal diagnosis of haemophilia*
Stage
Procedure
First trimester
Pre-implantation DNA diagnosis of embryonic blast cells
8-cell stage
Chorionic villus sampling
11–13 weeks
DNA analysis of fetal cells in maternal circulation
Transvaginal scanning for fetal gender
Second trimester
Amniocentesis
16 weeks
Cordocentesis—fetal umbilical cord sampling for clotting factor assay
Fetal gender determination
Third trimester (and at
delivery)
Fetal gender determination
Umbilical cord blood sampling
Adapted from (Kulkarni & Lusher 2001)
Three studies reported on the effectiveness of antenatal diagnosis for maternal carriers of
haemophilia A or B, or women with von Willebrand disease (Level IV evidence) (Table
47) (Daffos et al. 1988; Hoyer et al. 1985; Kadir et al. 1997).
The good quality study by Hoyer et al. 1985 (Level IV evidence) performed a foetoscopy,
which visualises the umbilical cord and samples the fetal blood supply by direct puncture
of the umbilical vein. Of 92 pregnancies, 80 foetoscopies obtained satisfactoryy fetal
blood samples. Of the 80 satisfactory samples, 51 pregnancies were diagnosed as normal.
However, post-natal testing revealed that one of these infants was positive for
haemophilia A (false negative=2%). The majority of the remaining 29 pregnancies, which
were diagnosed as positive for haemophilia A, were either terminated or miscarried. Postmortem analysis in 23 foetuses (82%) confirmed an abnormal result in all 23 cases.
One average quality case series (Level IV evidence) used chorionic villus sampling, the
current most commonly used method, to determine not only the sex of the foetus but
also whether a male foetus was affected by haemophilia A or B. The procedure is
performed at 11–13 weeks gestation and usually yields a result in 24–48 hours, but could
take as long as two weeks. Restriction fragment length polymorphisms or polymerase
chain reaction is applied to the sample. Sequencing of a known mutation may take 5–7
days. Kadir et al. 1997 recommend antenatal gender diagnosis by chorionic villus
sampling, but for those parents unwilling to undergo this procedure, ultrasound at 18
weeks gestation may be performed to ascertain gender.
Neither of the studies by Kadir et al. or Daffos et al. presented post-mortem results of
terminated foetuses to confirm the original diagnosis of an affected foetus.
Consequently, the accuracy of the antenatal testing is unknown in these studies.
y Samples contain a proportion of amniotic fluid, which dilutes the sample. Satisfactory samples = >10%
fetal blood; Unsatisfactory samples = <10% fetal blood
Use of recombinant and plasma-derived Factor VIII and IX
119
Table 47.
Effectiveness of antenatal diagnosis
Study
(Hoyer et al. 1985)
Level and quality
of evidence
Population
Level IV:
retrospective case
series
92 pregnancies
at risk of
haemophilia A
Fetal sampling
Foetoscopy: 80/92 (87%) satisfactory fetal samples
Normal
Quality score: 3/3
Selection bias
minimised
post-natal test
Follow-up adequate
Patients
representative
51/80 (63.8%)
1/51 2.0%) haemophilia A
3/51 (5.9%) fetal loss
47/51 (92.1%) normal males
46/47 (97.9%) confirmed normal
1/47 (2.1%) infant with reduced
levels of factor VIII
Haemophilia A 29/80 (36.2%)
26/29 (89.7%) terminated
1/29 (3.4%) haemophilia A carried to
term
2/29 (6.9%) fetal loss
post mortem a 23/28 (82.1%) abnormal result
confirmed
12/92 (13%) unsatisfactory samples
11/12 (91.7%) terminated
1/12 (8.3%) unaffected male
(Kadir et al. 1997)
Level IV:
retrospective case
series
Quality score: 2/3
Selection bias
minimised
32 women, 82
pregnancies
32/82 (39%) miscarriage or termination
24 haemophilia A
and 8
haemophilia B
obligate carriers
Chorionic villus sampling: 10/17 (58.8%)
4/10 (40%) females
2/10 (20%) unaffected males
4/10 (40%) affected males
2/4 (50%) terminated pregnancy
Follow-up adequate
17/48 (35%) had antenatal screening
Ultrasound gender screening: 7/17 (41.2%)
3/7 (42.9%) females
4/7 (57.1%) males
Patients
representative
Cordocentesis: 4 males
3/4 (75%) unaffected males
1/4 (25%) affected male, terminated
(Daffos et al. 1988)
Level IV: case
series
Quality score: 1/3
Selection bias likely
Follow-up adequate
Patients
representative
93 pregnant
women with
haemorrhagic
disorders
25/91 (27.5%) foetuses
24/25 (96%)
haemophilia A or B
terminated
Haemophilia A
35 obligate and
44 putative
carriers
Haemophilia B
8 women
obligate and 4
putative carriers
vWD
1 Type 1; 1 Type
2A
a
1/2 (50%) of women with vWD (Type 2A)
administered cryoprecipitate prior to fetal sampling
2/2 (100%) foetuses were affected by vWD
Only 23/28 terminations were made available for testing at post mortem; vWD = von Willebrand disease
Safety of antenatal diagnosis
Although there is minimal physical maternal risk, antenatal testing is not without physical
risk to the foetus. Only one good quality study reported on safety outcomes associated
with antenatal testing for haemophilia A (Level IV evidence) (Hoyer et al. 1985) (Table
120
Use of recombinant and plasma-derived Factor VIII and IX
48). There were five miscarriages (5%) within seven days of foetoscopy (of 92
pregnancies). In addition, there were five premature births in those pregnancies carried
to term (10%). However, authors report that this is equivalent to that seen in normal
pregnancies without foetoscopy.
Table 48.
Safety of antenatal diagnosis
Study
(Hoyer et al. 1985)
Level and quality
of evidence
Level IV:
retrospective case
series
Quality score: 3/3
Population
92 pregnancies
at risk of
haemophilia A
Complications
5/92 (5.4%) miscarriage within seven days of
foetoscopy
5/50 (10%) premature births in the pregnancies that
continued past foetoscopy
Selection bias
minimised
Follow-up adequate
Patients
representative
Overall, it appears that antenatal diagnosis by foetoscopy increases the risk of
termination to the foetus and may carry psychosocial consequences for the parent(s).
Although chorionic villus sampling is now used routinely for antenatal diagnosis, no
studies have directly compared the safety of foetoscopy and chorionic villus sampling
methods. Prior to invasive procedures, prophylaxis with factor concentrates may be
required (JBC FVIII/FIX working party & National Blood Authority 2004).
Haemophilia
Infants with severe haemophilia A or B may present with haemorrhage in the early
neonatal period (Kulkarni & Lusher 2001) and the delivery itself may be a time of
potential blood loss to the mother.
There were no studies that reported on the effectiveness of administering clotting factors
to pregnant women who were carriers for haemophilia A or B.
One study reported separate results for both haemophilia A and B (Greer et al. 1991) and
two large case series reported combined results for haemophilia A and B, for safety
outcomes associated with the delivery of infants (Kadir et al. 1997; Ljung et al. 1994)
(Level IV evidence) (Table 49).
Haemophilia A
Hormonal changes associated with pregnancy induce a rise in FVIII:C levels during the
second trimester, followed by a rapid drop after delivery. Therefore, carriers rarely
require prophylaxis for delivery, but they are at risk of bleeding during the first trimester
and 3–5 days after delivery (Economides et al. 1999). Two retrospective case series
(Level IV evidence) evaluated this occurrence.
It was reported that the majority of women who were carriers of haemophilia A had
increased levels of Factor VIII:C and von Willebrand antigen over the course of their
Use of recombinant and plasma-derived Factor VIII and IX
121
pregnancies, until they were far in excess of normal values at 28–36 weeks gestation
(normal values >45 IU/dl), with only two of 18 (11.1%) women failing to attain normal
levels of factor VIII:C (Greer et al. 1991). Similarly, Kadir et al. 1997 reported that levels
of factor VIII rose significantly in the second or third trimester compared to levels
measured in the first trimester in 37.5% of women who had measurements taken (p<
0.001).
No cases of primaryz postpartum haemorrhage were reported in the small case series
(Greer et al. 1991), however 8.8% of all pregnancies resulted in secondaryaa postpartum
haemorrhage, which was treated with cryoprecipitate or a blood transfusion.
Haemophilia B
Greer et al. 1991 reported that the majority of obligate carriers of haemophilia B have
normal levels of factor IX:C, therefore pregnant carriers of haemophilia B are unlikely to
experience major bleeding problems during labour. However, levels of factor VIII and
IX should be monitored throughout pregnancy because some women may not
experience an increase in factors during the course of their pregnancy and may be at risk
of prolonged postpartum bleeding (Level IV evidence). Kadir et al. 1997 reported that
there was no significant increase in levels of factor IX during pregnancy in 37.5% of
women who had measurements taken (no data given).
Greer et al. 1991 reported no cases of primary postpartum haemorrhage in their small
case series (nine term pregnancies) of obligate carriers of haemophilia B and only one
case of secondary postpartum haemorrhage, which required a blood transfusion. The
remaining eight pregnancies were treated with frozen fresh plasma as a precautionary
measure.
Haemophilia A and B
Kadir et al. 1997 reported that 36 (of 82) pregnancies in obligate carriers of haemophilia
A or B resulted in miscarriages, spontaneous abortions or terminations (Level IV
evidence). The remaining 46 pregnancies were predominantly normal vaginal deliveries
(70%). Primary (22%) and secondary postpartum haemorrhages (11%) required
treatment, including blood transfusion, or administration of frozen fresh plasma, or
infusion of factor VIII concentrates. A number of fetal or neonatal complications were
also reported, including fetal distress and bleeding complications. Kadir et al. 1997
recommended antenatal haemophilia diagnosis for the successful management of labour
or, at the very least, gender determination.
One large retrospective case series (Level IV evidence) of 117 deliveries reported a
significantly higher risk of fetal and neonatal complications in vaginal births with vacuum
extraction (64.7%) compared to normal vaginal delivery (8.7%) or caesarean section
(23%) (Ljung et al. 1994). In addition, 21% of neonates experienced a bleeding episode
z
Primary postpartum haemorrhage refers to bleeding >500ml during the 24 hours after delivery
Secondary postpartum haemorrhage refers to bleeding >500ml after the first 24 hours to six weeks after
delivery
aa
122
Use of recombinant and plasma-derived Factor VIII and IX
associated with procedures such as blood sampling and vitamin K injections. Ljung et al.
1994 recommended that the initial approach to delivery in a known haemophiliac should
be governed by obstetric factors and that vaginal delivery is not contraindicated.
Instrument delivery should be avoided (forceps and vacuum extraction), as should the
use of fetal scalp electrodes. Early recourse to caesarean is recommended when there is a
failure of labour to progress. Caesarean section for all cases where haemophilia is
expected is not likely to eliminate intracranial haemorrhage or other bleeding
manifestations.
Use of recombinant and plasma-derived Factor VIII and IX
123
Table 49.
Infant delivery and haemophilia
Study
Level and quality
of evidence
Population
(Greer et al. 1991)
Level IV:
retrospective case
series
18 obligate
carriers of
haemophilia A, 34
pregnancies
Quality score: 2/3
Selection bias
uncertain
Patients
representative
Level IV:
retrospective case
series
5 obligate carriers
of haemophilia B,
11 pregnancies
Quality score: 2/3
1 missed
miscarriage
(evacuation),
treated with FFP
Selection bias
uncertain
Follow-up
adequate
Vaginal deliveries: 23/34
Secondary PPH: 2/23 (8.7%)
Treatment: Evacuation of uterus and blood transfusion
Forceps deliveries: 7/34
Caesarean sections: 4/34
Secondary PPH: 1/4 (25%)
Treatment: cryoprecipitate for 7 days post-partum;
4 weeks later same patient, secondary PPH, received
cryoprecipitate and tranexamic acid
Follow-up
adequate
(Greer et al. 1991)
Outcomes
Vaginal deliveries: 5/11 (45.5%)
Perineal haematoma: 1/5 (20%)
Secondary PPH: 1/5 (20%)
Blood transfusion: 1/5 (20%)
Treatment: 4/5 (80%) FFP
Forceps deliveries: 2/11 (18.2%)
No PPH
Treatment: 2/2 (100%) FFP
Patients
representative
Caesarean sections: 2/11 (18.2%)
No PPH
Treatment: 2/2 (100%) FFP
(Kadir et al. 1997)
Level IV:
retrospective case
series
Quality score: 2/3
Selection bias
minimised
Follow-up
adequate
Patients
representative
32 women, 82
pregnancies
Miscarriage, spontaneous abortion, termination: 36/82
(43.9%)
24 haemophilia A
and 8
haemophilia B
obligate carriers
Vaginal deliveries: 32/46 (69.6%)
Mean gestation
39.2 weeks,
(range 34–42
weeks)
Primary PPH: 7/32 (21.9%)
Blood loss: 700–1500mls
Treatment: 2/7 (28.6%) FFP
2/7 (28.6%) factor VIII concentrate
1/7 (14.3%) recombinant factor VIII
Blood transfusion: 2/7 (28.6%)
Secondary PPH: 3/32 (9.4%)
Treatment: 1/3 (33.3%) FFP
1/3 (33.3%) factor VIII concentrate
Instrument deliveries: 6/46 (13%)
Primary PPH: 1/6 (16.7%)
Blood loss: 600mls
No treatment required
Caesarean sections: 8/46 (17.4%)
Primary PPH: 2/8 (25%)
Blood loss: 1200 and 1500 mls
Treatment: FFP + factor VIII concentrate; factor VIII
concentrate
Secondary PPH: 1/8 (12.5%)
No treatment required
124
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 49 (cont.)
Study
Level and quality
of evidence
(Ljung et al. 1994)
Sweden
Level IV: retrospective case
series
Quality score: 2/3
Selection bias
minimised
Follow-up
adequate
Patients
representative
Population
Outcomes
117 pregnant
women, 117
deliveries of
haemophilic
children
Bleeding episodes: 23/117 (19.7%) associated with
delivery
24/117 (20.5%) neonates (<2 weeks of age) had bleeding
episodes associated with procedures eg blood sampling,
vitamin K injections, bleeding post-surgery.
101 haemophilia
A (77 severe, 24
moderate)
6/117 (5.1%) neonates required blood transfusion
16 haemophilia B
(12 severe, 4
moderate)
Vaginal deliveries: 104/117 (88.9%)
Primary PPH: 3/104 (2.9%)
Fetal/neonatal complications: 9/104 (8.7%)
Subgaleal /cephalic 1/104 (0.9%)
Intracranial 2/104 (1.9%)
Umbilical 3/104 (2.9%)
Retro-orbital 1/104 (0.9%)
Oral 1/104 (0.9%)
Haematuria 1/104 (0.9%)
Vacuum extraction vaginal delivery: 17/104 (16.3%)
Fetal/neonatal complications: 11/17 (64.7%)
Subgaleal /cephalic 10/17 (58.9%)
Intracranial 1/17 (5.9%)
Umbilical 0/17 (0.0%)
Retro-orbital 0/17 (0.0%)
Oral 0/17 (0.0%)
Haematuria 0/17 (0.0%)
Caesarean sections: 13/117 (11.1%)
Primary PPH: 1/13 (7.7%)
Fetal/neonatal complications: 3/13 (23%)
Subgaleal /cephalic 1/13 (7.7%)
Intracranial 1/13 (7.7%)
Umbilical 1/13 (7.7%)
Retro-orbital 0/13 (0.0%)
Oral 0/13 (0.0%)
Haematuria 0/13 (0.0%)
FFP = frozen fresh plasma, PPH = post-partum haemorrhage
von Willebrand disease
For women with von Willebrand disease (vWD), particularly severe vWD, the delivery of
a child may result in severe bleeding unless treated appropriately. All four studies that
met the inclusion criteria were low-level evidence (Level IV).
Three poor–average quality studies reported on safety outcomes associated with the
pregnancy and delivery of infants in women affected by vWD (Chediak et al. 1986; Greer
et al. 1991) (Table 50).
All three studies noted that the levels of factor VIII, von Willebrand antigen and von
Willebrand factor activity increased significantly during gestation (Ljung et al. 1994,
p=0.0001), peaking in the third trimester (no data given).
Vaginal bleeding resulted in the miscarriage or elective termination of a relatively high
proportion of pregnancies in the study by Kadir et al. 1998, with 36% continuing to full
term. Complications of pregnancy loss included excessive bleeding (10%) and secondary
Use of recombinant and plasma-derived Factor VIII and IX
125
haemorrhage (10%), which required blood transfusion and/or cryoprecipitate or factor
concentrate. The majority of births in all three studies were normal vaginal delivery.
All studies reported that postpartum haemorrhage was a common outcome for women
with vWD. Primary postpartum haemorrhage was reported in all three studies, with rates
ranging from 19–27% of deliveries, resulting in severe bleeding which required treatment
either with cryoprecipitate, whole blood or fresh plasma. Secondary postpartum
haemorrhage was reported by two of the three studies and occurred in 20–36% of
deliveries. Chediak et al. 1986 reported that postpartum haemorrhage tended to occur in
women with more severe type of vWD. However, the number of women enrolled in this
study was too small to make any firm conclusions. The larger retrospective case series by
Kadir et al. 1998 did not confirm the von Willebrand status of women affected by
postpartum haemorrhage. Greer et al. 1991 noted that the high rate of postpartum
haemorrhage was likely to be associated with the increase in factor VIII during
pregnancy, combined with the rapid fall during delivery, especially in women with Type 2
and Type 3 vWD. In patients with Type 1 vWD, a rise in factor VIII to >50 IU/dl
appears to be sufficient to allow an uncomplicated vaginal delivery. Greer et al. 1991
recommended that all women with Type 2 or 3 vWD, and those Type 1 women who fail
to reach optimal factor VIII:C levels (50 IU/dl), be treated immediately postpartum with
blood products containing von Willebrand factor. In addition, women who deliver by
caesarean should be administered blood products.
The study by Kadir et al. 1998 reported 4% of infants, born to women with vWD,
experienced bleeding episodes immediately after delivery and recommended that invasive
fetal monitoring techniques such as scalp electrodes or fetal blood sampling should be
avoided.
Chediak et al. 1986 recommended that the delivery of infants by women affected by
vWD should be assessed on an individual basis.
In addition, all the women enrolled in the case series reported by Greer et al. 1991 had
been referred to the clinic for menorrhagia and reported prolonged menses, ranging in
duration from five to 14 days. Although vWD is not a common disorder, it is the most
common inherited bleeding disorder and Greer et al. recommend that it should be
considered in the assessment of women referred for menorrhagia and for their future
management during pregnancy.
126
Use of recombinant and plasma-derived Factor VIII and IX
Table 50.
Infant delivery and von Willebrand disease
Study
(Schulman et al.
1991)
Level and quality of
evidence
Level IV: case series
Quality score: 3/3
Measurement bias
minimised
Selection bias
minimised
Population
Outcomes
370 patients with
disorders of primary
haemostasis. 133
vWD, 237 platelet
function defects,
aged 3–80 years
DDAVP (0.2–0.3 µg/kg, route of administration
not specified)
8 women with von
Willebrand, 14
pregnancies
Vaginal deliveries: 12/14
When given before labour in one patient, labour
ceased and had to be induced.
Follow-up adequate
(Greer et al. 1991)
Level IV: retrospective
case series
Quality score: 2/3
Primary PPH: 1/12
Treatment: blood transfusion
Selection bias
uncertain
Secondary PPH: 4/12
Treatment: Cryoprecipitate, blood transfusion
Follow-up adequate
Maternal complications: 1/12
Threatened miscarriage
Patients
representative
Forceps deliveries: 1/14
Primary PPH: 1/1
Treatment: cryoprecipitate, blood transfusion
Secondary PPH: 1/1
Treatment: cryoprecipitate, blood transfusion
Caesarean sections: 1/14
Treatment: cryoprecipitate
(Kadir et al. 1998)
Level IV: retrospective
case series
Quality score: 2/3
Selection bias
uncertain
Follow-up adequate
Patients
representative
(Chediak et al.
1986)
Level IV: case series
Quality score: 1/3
Selection bias likely
Follow-up adequate
31 women, 84
pregnancies, 85
foetuses
27 Type 1; 2 Type
2; 2 Type 3
Live birth 55/85 (64.7%)
Miscarriage or termination: 30/84 (35.7%)
Fetal or neonatal complications: 2/55 (3.6%)
Mean gestation 40
weeks (range 34–42
weeks)
Caesarean sections: 5/54 (9.3%)
Vaginal deliveries: 43/54 (79.6%)
Forceps deliveries: 6/54 (11.1%)
Blood loss: 750->2000 mls
Treatment: pdFVIII/vWF; FFP; blood transfusion
Six women, eight
deliveries, 11
pregnancies (3
spontaneous
abortions)
Patients
representative
Fetal or neonatal complications: Data for only 7/8
(87.5%) deliveries
5/7 (71.4%) affected
Vaginal deliveries: 3/8
Primary PPH: 2/3
Severe vaginal bleeding, massive lumbar bleeding
Treatment: Whole blood, fresh plasma,
cryoprecipitate
Caesarean sections: 5/8
Primary PPH: 1/5
Vaginal bleeding
Treatment: Cryoprecipitate
Maternal complications: 2/5
placenta praevia, membrane rupture, water
intoxification after desmopressin
Treatment: cryoprecipitate before delivery,
curettage, postpartum desmopressin
DDAVP = 1-D-deamino-8D-arginine vasopressin; FFP = fresh frozen plasma; pdFVIII/vWF = von Willebrand factor rich concentrate; PPH =
postpartum haemorrhage
Use of recombinant and plasma-derived Factor VIII and IX
127
No available studies concerning the delivery of infants in other rare bleeding disorders
met the inclusion criteria for an evaluation of effectiveness or safety.
A range of other issues that may impact on the care of newborns with haemophilia A or
B was reported in the literature. The recommendations relating to these issues were
based on case reports and expert opinion, a lower level of evidence than that included for
evaluation in this systematic review.
The early neonatal period is a time of high risk for the newborn with haemophilia. The
pattern of bleeding in neonates differs markedly from that seen in older children with
haemophilia (Kulkarni & Lusher 2001). From a narrative review of histories of 349
newborns with haemophilia, the most frequent bleeding events occurred following
circumcision (30%—most of which occurred prior to 1970), intracranial haemorrhage
(27%), puncture bleeds (16%)—such as heel sticks, venipuncture, and vitamin K
injections—and subgaleal or cephalohaematoma (13%) (Kulkarni & Lusher 2001). Joint
bleeds, the main bleeding event in older children, represent one per cent of bleeding
episodes in neonates. Since these data were gathered from published case reports, they
are prone to publication bias. However, they provide an indication of the difference in
the patterns of bleeding presented in newborns and older children or adults.
Expert opinion suggests that pregnancy in women with rare coagulation disorders should
be managed by an obstetrician in collaboration with a specialist haematologist (Chalmers
2004; Giangrande 1998; Kulkarni & Lusher 2001). Recommendations include:
immunisation against hepatitis B, as a safeguard should blood products be required at
delivery; if the use of forceps is necessary to avoid prolonged labour, they should be
applied with great care, by avoiding rotation of the head in midcavity and limiting their
use to low forceps delivery when the head is deeply engaged in the pelvis. Recombinant
coagulation factor may be required after forceps delivery in haemophilia newborns;
newborns with mild haemophilia and a normal screening test (prothrombin time or
activated partial thromboplastin time)bb, may still be at risk of a bleed, such as intracranial
haemorrhage in the first few days after birth. Therefore, factor assays should be
performed on cord or peripheral samples and repeated at six months; since factor IX
levels in mild haemophilia B may overlap with normal low FIX at birth, repeat testing
should be performed at six months; all infants with intracranial haemorrhages should be
evaluated for a bleeding disorder, as >30 per cent of all newborns with haemophilia have
no family history of haemophilia; in cases of haemorrhage, rFVIII or rFIX should be
administered to provide immediate haemostasis. If diagnosis of haemophilia is
unconfirmed, 15–20 ml/kg fresh frozen plasma may be considered for acute
haemorrhage; and any necessary injections for the newborn should be performed with
extra care and followed by local application of pressure applied for five minutes with
close observation of the site for 24 hours. If umbilical stump bleeding occurs, infants
should be tested for factor II, X or factor XIII deficiency (Kasper 2004).
bb Prothrombin time (PT) measures the time required for blood to clot and may be used to measure the
presence and activity of factors I, II, V, VII and X. PT is lengthened by quantitative or functional
reduction in clotting factors
Partial thromboplastin time (PTT) measures the time needed for blood to clot and may be used to screen
for factor deficiencies. PTT is increased above normal (25–41 seconds) with heparin, warfarin,
thrombolytics, clotting factor deficiency, liver disease, vitamin K deficiency and disseminated intravascular
coagulation
128
Use of recombinant and plasma-derived Factor VIII and IX
Evidence-based clinical practice guidelines
On the basis of available evidence, pregnancy and infant delivery in haemophilia carriers and women
with von Willebrand disease should be managed as follows (Level IV evidence).
Antenatal care
•
Genetic counselling should be accessible to all haemophilia carriers and women with von
Willebrand disease covering the potential risks (to the foetus and mother) of both antenatal testing
and complications during pregnancy and delivery.
•
Antenatal testing options depend on the stage of pregnancy. All procedures carry risks—chorionic
villus sampling is the preferred method. Factor replacement may be required for invasive
procedures (expert opinion).
•
Haemostatic support is rarely necessary for haemophilia A carriers, but may be required for
invasive tests. However, since levels of FVIII rise during normal pregnancy, from the second
trimester, and drop immediately after delivery, the risk of haemorrhage is higher during the first
trimester and 3–5 days after delivery. Therefore, FVIII:C levels should be monitored.
•
Since levels of FIX remain unchanged throughout pregnancy, women with low baseline FIX may
require rFIX to control haemorrhage. FIX:C should be measured soon after pregnancy is confirmed
and monitored in the third trimester and postpartum.
Delivery of infants
•
Vaginal delivery is preferred for all infants with haemophilia or suspected haemophilia unless
obstetric factors indicate caesarean section. If necessary, early recourse to caesarean is the
preferred alternative, with monitoring of factor levels and an available supply of appropriate
recombinant factor concentrate.
•
The use of invasive fetal monitoring, such as scalp electrodes, instrument deliveries and long
labours should be avoided in affected male foetuses, or if fetal sex or coagulation status is
unknown. Vacuum (Ventouse) extraction is contraindicated due to the higher risk of fetal
complications.
Postpartum and postnatal period–Newborn
•
Cord or peripheral blood samples should be taken from male infants with suspected bleeding
disorders to ascertain coagulation factor status.
•
Heel sticks and venipuncture should be avoided if possible and vitamin K injections should be
delayed until diagnosis of haemophilia is excluded. Oral vitamin K may be used as the preferred
route for prophylaxis in confirmed haemophilia infants.
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
129
(cont.)
Postpartum and postnatal period—Mother
•
Women may develop inhibitors to FVIII or FIX postpartum. See Acquired haemophilia.
•
Although prophylactic use of DDAVP should be avoided during labour, women with a mild form of
von Willebrand disease (Type 1) may require DDAVP to increase and maintain factor levels in the
first 3–4 days after delivery. See section on DDAVP.
•
Patients with Type 2 or 3 von Willebrand disease and those with Type 1 who fail to reach optimal
FVIII:C levels may be treated with pdFVIII/vWF concentrates postpartum.
130
Use of recombinant and plasma-derived Factor VIII and IX
Other Aspects of Management
There is a range of management issues that should be considered in the treatment of
patients with coagulation disorders. However, there is very little published evidence on
which to base recommendations. The following section on comprehensive care and
management of bleeding episodes is largely informed by low-level evidence and expert
opinion.
Comprehensive care
The optimal management of patients includes more than treating or preventing acute
bleeding episodes. It should include the management of disease-related joint and muscle
damage and address psychosocial needs such as education, employment, psychology and
psychiatry services. The aim of disease management is to optimise quality of life by
minimising disease and treatment-associated morbidity and mortality. These goals can be
attained through comprehensive care, including home therapy, and access to safer
products (Association of Hemophilia Clinic Directors of Canada 1995a).
Specialist haemophilia centres are best equipped to provide comprehensive
interdisciplinary care for patients with blood coagulation disorders. Therefore, all aspects
of disease management, including rheumatology, orthopaedic surgery, dentistry, clinical
genetics, infectious diseases, physiotherapy and gynaecology should be managed in
consultation with a haemophilia specialist. One good quality retrospective cohort study
of 2950 patients with haemophilia (Level III-2 evidence) found that those patients whose
treatment was managed through a haemophilia treatment centre had a significantly
decreased risk of death than those treated elsewhere, even after adjustment for other
associations through multivariate analysis [RR=0.6, 95% CI 0.5, 0.8; p=0.002] (Soucie et.
al 2000).
Haemophilia and von Willebrand disease have a large impact on families in many ways,
and parents, spouses and siblings can participate actively in patient care. It is essential
that families be well informed about the diseases and their implications, including
treatment strategies. Parents and persons with haemophilia are often very medically
sophisticated and knowledgeable about the disease, so should be involved in decisionmaking processes about treatment strategies.
The extended family may require genetic counselling and, when appropriate, be offered
carrier testing and antenatal diagnosis (Association of Hemophilia Clinic Directors of
Canada 1995a). Mutation testing of affected individuals is crucial to provide accurate
diagnosis of carrier status (expert opinion, AHCDO Executive Committee 2005).
As a result of the transmission of viral infections, such as HIV, hepatitis B and C, the
need for counselling of both the patient and the family has increased, prompting greater
need for education about safe sexual practices for those who are infected (Association of
Hemophilia Clinic Directors of Canada 1995a).
Home therapy allows patients with severe or moderate haemophilia to have immediate
access to treatment in the event of a haemorrhage. This may result in decreased medical
costs, improved quality of life, and decreased amount of time spent in hospital and away
from school and employment (Valentino et alr 2004). Soucie et al. 2001 studied the
Use of recombinant and plasma-derived Factor VIII and IX
131
relationship between home therapy (receiving factor infusions outside a medical setting)
and haemorrhagic bleeding complications over a four year period in a good quality
retrospective cohort study (Level III-2 evidence). Patients receiving home therapy were
20% less likely to have a haemorrhagic bleeding complication than those who did not use
home therapy (RR= 0.8, 95% CI 0.7, 0.9; p≤0.05). After adjusting for all other
determinants—such as disease severity, ethnicity, employment status and health
insurance—patients using home therapy and consulting a haemophilia treatment centre
had the highest probability of avoiding a haemorrhagic bleeding complication during
follow-up. Those patients using neither had the lowest probability of avoiding a
haemorrhagic bleeding complication during follow-up, whereas patients using either
home therapy or a haemophilia treatment centre had intermediate probability of avoiding
bleeding complications.
Central venous access devices may facilitate the use of home treatment in those less than
four years of age, and detailed education pertaining to the essential procedures required
to avoid infections and other complications should be provided to patients and their
families (Valentino et al. 2004).
Management of bleeding episodes
When a patient with haemophilia presents with a bleeding episode, the administration of
factor replacement should be the first course of action, particularly when the bleeding
occurs in the head, neck, chest or abdomen and may be life threatening. Once infused,
diagnostics and examinations may begin. For most acute bleeds, once factor concentrates
have been administered, discharge instructions should follow the rule of RICE (rest, ice,
compression, elevation), taking weight off through use of crutches etc. and, in the event
of a joint bleed, supporting the joint through the use of splints and other mobility aids.
Issues in treating young patients
The feasibility of prophylaxis or tolerisation treatments for patients with haemophilia
depends on the availability of adequate venous access for use in home therapy. In young
children, repeated access to peripheral veins may be difficult. To overcome this problem,
two main methods have been developed: 1) central venous access devices—either fully
implanted or external; or 2) arteriovenous fistulae, an alternative internal vascular access
device for factor infusion. The decision to introduce a central venous catheter should
take into account the combined social and medical indications (access to a peripheral
vein, feasibility of home treatment) while balancing the potential risks (infections,
thrombosis or other rare complications). It has been suggested that implantation of
central venous catheters on psychological grounds alone should be discouraged and
children who are afraid of venipuncture should be assisted by other means (Ljung 2004).
Safety of central venous access in haemophilia
The best available information on the safety of central venous access devices (CVADs)
was obtained from a systematic review of 48 studies (Level IV evidence) and 2704
patients covering the benefits and risks of both fully implanted and external devices in
haemophilia patients. The results were pooled where possible, and a total of 1190
infections were reported (40% of CVADs and 44% of patients), giving a pooled
132
Use of recombinant and plasma-derived Factor VIII and IX
incidence of 0.66 per 1000 CVAD days. The most common types of infection were due
to Staphylococcus epidermidis or Staphylococcus aureus, which showed that the predominant
source of catheter-related infections were from the flora of the patient’s skin. The three
independent risk factors for infection were age, inhibitors and type of CVAD. Children
over six years of age were 46% more likely to develop infections compared to those aged
2–6 years. Those with inhibitors were 67% more likely to develop infection than those
without. Meanwhile, the incidence of infections in those with fully implanted CVADs
was only 31% of the rate of those with external CVADs. The pooled incidence of
thrombotic complications from CVADs was 0.06 per 1000 CVAD days (1.8% of
CVADs and 2.0% of patients) and when a multivariate analysis was performed, neither
age nor presence of inhibitors was found to affect the incidence of thrombosis (Level IV
evidence) (Valentino et al. 2004). More serious complications included pinch-off
syndrome (which occurs when the catheter becomes blocked, due to compression
between the first rib and the clavicle), port erosion (where the skin overlying the septum
deteriorates), or when the child outgrew the catheter and the tip became separated from
the optimal vessel.
In order to reduce infections associated with CVAD use, strict adherence to hand
washing and aseptic technique remains vital. Adequate initial training and frequent review
and reinforcement of procedures may reduce infection rates (Valentino et al. 2004).
In young patients where the peripheral vein is not accessible CVADs are a valuable tool
for assisting in the administration of factor concentrates, which may avert life-threatening
haemorrhages or prevent development of arthropathy. However, the risk of infections
and other rare complications means that health professionals should periodically review
the need for a CVAD and peripheral veins should be used where possible (Valentino et
al. 2004).
Safety of internal arteriovenous fistulae
One prospective case series examined the use of internal arteriovenous fistulae as an
alternative to central venous access devices. From a series of 27 patients and 31 devices,
no infectious complications were noted. However, the authors found that five out of 31
arteriovenous fistulae did not develop or maintain sufficient blood flow for long-term
factor concentrate infusion. Five patients had complications, such as bleeding,
thrombosis, and symptoms of distal ischaemia. Overall, 84% of devices with successful
access were deemed suitable for long-term use (Level IV evidence) (Santagostino et al.
2003). Currently, there is insufficient evidence to make recommendations on the use of
internal arteriovenous fistulae.
Evidence-based clinical practice guidelines
•
On the basis of the evidence, central venous access devices assist in administering factor
concentrates in children. However, adequate aseptic technique should be taught and frequently
reviewed in order to avoid infection (Level IV evidence).
•
The evidence suggests that external devices should be avoided due to much higher rates of
infection than fully implanted devices (Level IV evidence).
Use of recombinant and plasma-derived Factor VIII and IX
133
Summary and Conclusions
The previous Australian clinical practice guidelines for the treatment of haemophilia A,
B, von Willebrand disease and other rare bleeding disorders are based on the consensus
of experts in the field of coagulation disorders (See Appendix G). These guidelines were
modified by the Haemophilia Foundation Australia Medical Advisory Panel in March
2000 from existing guidelines developed by the United Kingdom Haemophilia Centre
Directors Organisation Executive Committee (Haemophilia Foundation Australia
Medical Advisory Panel 2000).
The consensus guidelines were limited to:
•
Selection of products
•
Safety data
•
Recommendations about informing patients and consent, vaccination against
hepatitis A and B, risk reduction, use of DDAVP, tranexamic acid and aminocaproic
acid
•
Specific recommendations about products most suitable for haemophilia A, B, von
Willebrand disease, deficiencies in factors II, V, VII, X, XI, XIII, or fibrinogen, and
future treatment of hereditary coagulation disorders.
Areas that consensus guidelines did not address included:
•
Recommendations for the use of recombinant products for all patients with
haemophilia A and B
•
Tolerisation
•
Treatment of patients with inhibitors
•
Prophylaxis
•
Infant delivery
•
Acquired haemophilia
•
Surgical and dental procedures
•
Other management issues.
The new evidence-based guidelines have attempted to address these areas and provide
recommendations based on the highest level of available evidence. Where empirical
evidence was not available, recommendations follow those in the previous guidelines
and/or consensus-based expert opinion. The evaluation of studies for this review was
performed in the context of the newly introduced policy to provide access to
recombinant products for all people with haemophilia A or B.
134
Use of recombinant and plasma-derived Factor VIII and IX
In the absence of good quality comparative studies in several sections of this review, the
recommendations for the treatment and management of haemophilia A, B, vWD and
other rare coagulation disorders have been based on a large body of low level evidence
that is heterogeneous in nature and of relatively poor quality.
Research question 1
What is best practice for the treatment of acute bleeding episodes in patients with
haemophilia A, B, vWD and other rare coagulation disorders?
Haemophilia A, without inhibitors
The best available evidence indicates that recombinant FVIII is the treatment of choice
for patients without inhibitors to FVIII (Level II evidence for effectiveness; Level III-2
for safety). Inhibitor testing, particularly in young children and/or those with severe
haemophilia, should be performed no earlier than three days after initial factor
concentrate administration, or when the expected response is absent, with re-testing at
regular intervals (Level IV evidence).
Patients with mild–moderate haemophilia A may be treated using DDAVP, at a dose of
0.3 µg/kg diluted in 50ml of 0.9% saline and infused over ≥30 minutes. DDAVP may be
administered intravenously or subcutaneously with comparable results (Level IV
evidence). DDAVP may be administered once every 24 hours, becoming less effective
after repeated doses (Level IV evidence). Predose monitoring of electrolyte
concentrations is recommended if DDAVP is administered more than once in 24 hours
(expert opinion). A test dose of DDAVP and FVIII/vWF assay should be performed to
demonstrate efficacy (Level IV evidence). Caution should be taken to restrict fluid intake
during DDAVP treatment to prevent fluid overload (Level IV evidence). If DDAVP is
given more than once in a 24 hour period, predose monitoring of electrolyte
concentrations is recommended (expert opinion). DDAVP should be used with caution
in the elderly; and it is not recommended in those with arteriovascular disease and in
young children (<2 years) (expert opinion).
Haemophilia B, without inhibitors
The best available evidence indicates that recombinant FIX is safe and effective for the
treatment of bleeding episodes in haemophilia B patients without inhibitors (Level IV
evidence for safety and effectiveness). Due to large inter-patient variability, individual
dosing regimens should be monitored by FIX recovery assays (Level II evidence).
Compared to plasma-derived FIX, recombinant FIX dosage should be increased 1.6
times for patients aged ≤15 years and 1.2 times for patients aged 16 years and over (Level
III-2 evidence). Since there is a risk of anaphylaxis occurring within the first 50 FIX
exposure days, patients should be closely monitored (expert opinion).
Von Willebrand disease
Patients with mild vWD may be treated using DDAVP in the manner described above
for patients with mild–moderate haemophilia A. DDAVP should be used only as an
adjunct to factor replacement therapy in patients with severe vWD and not as the
primary treatment (Level II evidence). Patients unresponsive to (or contraindicated for)
DDAVP should receive pdFVIII/vWF concentrate, which may be administered every 8–
12 hours (Level IV evidence).
Use of recombinant and plasma-derived Factor VIII and IX
135
Research question 2
What is best practice for the prophylactic treatment of patients with haemophilia A, B or
von Willebrand disease?
Based on Level II evidence, prophylaxis is recommended for haemophilia patients
without inhibitors. Prophylaxis with factor concentrates should be initiated after bleeding
episodes have commenced, rather than at diagnosis, due to the possible increased risk of
inhibitor development associated with administration of factor concentrates in very
young children (Level IV evidence).
Central venous access devices may assist in the regular infusion of factor concentrates in
children. However, adequate aseptic technique should be taught and frequently reviewed
to avoid infection (Level IV evidence). External venous access devices should be avoided
due to the higher rates of infection compared to fully implanted devices (Level IV
evidence).
Haemophilia A, without inhibitors
Recombinant FVIII is the treatment of choice for prophylaxis in patients without
inhibitors to FVIII (Level IV evidence for safety and effectiveness).
Haemophilia B, without inhibitors
Recombinant FIX is safe and effective for prophylaxis in haemophilia B patients without
inhibitors (Level II evidence for effectiveness; Level IV evidence for safety). As discussed
in Research question 1, dosing regimens should take into account the lower rFIX
recovery compared to pdFIX and inter-patient variability (Level II evidence).
Von Willebrand disease
There was insufficient evidence to determine the effectiveness of intranasal DDAVP as
prophylaxis for the prevention of menorrhagia in women with von Willebrand disease
(Level II evidence).
Research question 3
What is the best method for treating bleeding episodes in patients with inhibitors to
factor VIII or IX?
Haemophilia A, with inhibitors to FVIII
Recombinant FVIIa is the treatment of choice for acute bleeding episodes in patients
with high titre and/or high responding inhibitors to FVIII (Level II evidence for
effectiveness; Level IV evidence for safety). Recombinant FVIIa may be infused as a
bolus dose of 90 µg/kg for adults (Level IV evidence). Doses up to 200–250 µg/kg may
be required for children (expert opinion). Activated prothrombin complex concentrates
(aPCCs) may be considered for controlling mild–severe bleeding in patients with high
titre inhibitors. However, patients should be closely monitored for adverse reactions,
which, though rare, may be serious. The maximum daily dose of FEIBA should not
exceed 200 IU/kg (Level II evidence). Plasmapheresis (with or without
immunoadsorption) may be used to reduce inhibitors in high titre/high responders
before infusion with FVIII to control bleeding. However, patients should be monitored
for potential anaphylactic reactions (Level IV evidence). There is insufficient evidence to
136
Use of recombinant and plasma-derived Factor VIII and IX
recommend the use of high dose FVIII concentrates for the treatment of acute bleeding
episodes in patients with inhibitors to FVIII. Immunosuppression therapy with
cyclophosphamide to reduce inhibitors is not recommended for treating acute bleeding
episodes (Level IV evidence).
Haemophilia B, with inhibitors to FIX
Recombinant FVIIa is the preferred treatment for acute bleeding episodes in patients
with high titre and/or high responding inhibitors to FIX (Level II evidence for
effectiveness; Level IV evidence for safety). APCCs also control mild–severe bleeding in
patients with high titre inhibitors. However, patients should be closely monitored for
adverse reactions (Level IV evidence).
Acquired haemophilia
Bleeding episodes in patients with acquired haemophilia require prompt treatment (Level
IV evidence).
Inhibitors <5 BU: DDAVP should be used at the dose recommended for von
Willebrand disease (Research question 1). Plasma-derived factor VIII concentrate may be
used at a dose of 20 IU/kg for each BU of inhibitor plus an additional 40 IU and tested
after 15 minutes. If response is poor, another bolus injection may be given (Level IV
evidence).
Inhibitors >5 BU: Activated prothrombin complex concentrates (aPCCs) may be
administered at a dose of 50–200 IU/kg/day in divided doses (Level IV evidence).
Activated recombinant FVII (rFVIIa) may be administered at a dose of 90 µg/kg every
2–6 hours until the bleeding stops (Level IV evidence). Due to the risk of thrombotic
events, patients should be closely monitored after treatment (Level IV evidence).
Research question 4
What is best practice for the implementation of tolerisation procedures for patients with
inhibitors to factor VIII or IX?
Haemophilia A, with inhibitors to FVIII
There is no consensus on tolerisation protocols in Australia. Protocols used range from
25 IU/kg three times per week, to 100 IU/kg/day (JBC FVIII/FIX working party &
National Blood Authority 2004). When factor concentrates are used, recombinant factors
are preferred rather than plasma-derived factors due to their higher safety profile
regarding transmission of blood-borne agents (Level IV evidence). The Bonn protocol
may be considered for young patients (Level IV evidence). Treatment is more likely to be
successful in patients with low pre-treatment inhibitor titres; and when there is less time
between the development and treatment of inhibitors (Level IV evidence). The Malmö
protocol may be considered when patients have long-standing inhibitors (Level IV
evidence).
Haemophilia B, with inhibitors to FIX
Recombinant FIX is the preferred treatment for tolerisation protocols in patients with
inhibitors to FIX (Level IV evidence). Close monitoring is recommended to prevent
anaphylactic reactions (expert opinion).
Use of recombinant and plasma-derived Factor VIII and IX
137
Acquired haemophilia
Patients with acquired haemophilia should be treated for the underlying disorder before
deciding on strategies to eliminate inhibitors (Level IV evidence).
Postpartum: Patients may be monitored, without treatment, as inhibitors most frequently
disappear spontaneously. Alternatively, immunosuppressive therapy may be considered
(Level IV evidence).
Drug-related: The drug should be withdrawn to allow spontaneous disappearance of
inhibitors. Alternatively, immunosuppressive therapy may be considered (Level IV
evidence).
Autoimmune disease: Immunosuppressive therapy should be given, as inhibitors are
unlikely to disappear spontaneously (Level IV evidence).
Malignant neoplasm: Patients should be treated for the primary malignancy.
Immunosuppressive therapy should then be given (Level IV evidence).
Idiopathic: Patients should be given immunosuppressive therapy (Level IV evidence).
Research question 5
What is the best method of managing patients with haemophilia A, B, von Willebrand
disease and other rare coagulation disorders undergoing surgical and dental procedures?
Consensus-based recommendations for the management of patients undergoing surgery
(Association of Hemophilia Clinic Directors of Canada 1995a) or dental procedures
(Stubbs & Lloyd 2001) have been outlined in detail (Table 37 and Appendix G). The
available evidence base that was evaluated in this review is consistent with the
recommendations in these existing guidelines.
Based on the evidence evaluated in the current review, the following management
approaches are recommended:
Haemophilia A or B, without inhibitors
Recombinant factor VIII is safe (Level III-2 evidence) and effective (Level IV evidence)
for haemophilia A patients and recombinant factor IX is safe and effective (Level IV
evidence) for haemophilia B patients, before or after surgery as required. Tranexamic acid
may be used as secondary prophylaxis for surgical or dental procedures (Level IV
evidence).
Haemophilia A or B, with inhibitors
Recombinant FVIIa may be considered the first line of treatment for dental and other
surgical procedures in patients with high titre and/or high responder inhibitors. Evidence
for the use of recombinant FVIII/FIX concentrates during surgery or dental procedures
is currently awaited. Factor VIII concentrate may be considered for use during surgery in
patients with low titre, low-responding inhibitors to FVIII, but should be avoided in
patients with high-responding inhibitors (Level IV evidence).
138
Use of recombinant and plasma-derived Factor VIII and IX
Von Willebrand disease
DDAVP, which may be used in conjunction with topical therapies such as fibrin glue and
tranexamic acid, may be considered for use prior to dental extractions or surgery in
patients who respond to DDAVP. A test dose is recommended to determine
responsiveness (Level IV evidence). Plasma-derived FVIII/vWF concentrates may be
used to control bleeding in patients who do not respond to DDAVP (Level IV evidence).
Research question 6
What is best practice in the delivery of infants with haemophilia A or B and the
management of infant delivery in women with von Willebrand disease and other rare
coagulation disorders? Unless stated, all recommendations are based on Level IV
evidence.
Antenatal care
Counselling pertaining to the potential risks of antenatal testing and the potential
complications during pregnancy and delivery should be accessible to all haemophilia
carriers and women with von Willebrand disease. If antenatal testing is accepted,
chorionic villus sampling is the preferred method. Factor concentrates may be required
for invasive procedures. FIX levels in haemophilia B carriers and FVIII levels in
haemophilia A carriers should be monitored, particularly in the third trimester and
postpartum.
Delivery of infants
Vaginal delivery is the recommended mode of delivery, unless obstetric factors indicate
caesarean section. Invasive fetal monitoring, such as scalp electrodes, instrument
deliveries and long labours should be avoided for affected male infants. Vacuum
extraction delivery is contraindicated. Use of DDAVP in women with von Willebrand
disease is not recommended during labour.
Postpartum and postnatal care
Cord or peripheral blood samples, to ascertain coagulation factor status, should be taken
from male infants of haemophilia carriers and infants with suspected bleeding disorders.
All infants with intracranial haemorrhage should be evaluated for the presence of a
bleeding disorder, even where there is no family history of haemophilia. DDAVP is
contraindicated in neonates. Heel sticks and venipuncture should be avoided if possible,
other than to assess coagulation factor status. Intramuscular vitamin K is associated with
bleeding in infacts with haemophilia. Oral vitamin K is available as an alternative. Fibrin
glue may be used in conjunction with factor concentrates to achieve haemostasis if
circumcision is performed (Level IV evidence). DDAVP may be used for prophylaxis
during the first 3–4 days after delivery in women with mild Type 1 von Willebrand
disease to increase and maintain factor levels. All Type 2 and 3 von Willebrand disease
and Type 1 patients who fail to reach optimal FVIII:C levels may be treated with
pdFVIII/vWF concentrates postpartum (Level IV evidence).
Use of recombinant and plasma-derived Factor VIII and IX
139
Areas for further research
Participation in international and national research projects
Although the published literature pertaining to haemophilia and von Willebrand disease
comprises a large quantity of lower level research (uncontrolled before-and-after studies,
case series and case reports), there is a dearth of good quality comparative studies that
minimise bias and confounding. With regard to the other rare bleeding disorders
described in this review, the evidence base is particularly sparse. If future treatments are
to be based on reliable evidence, better-designed higher-level studies are required to
investigate the safety and effectiveness not only of haemostatic products, but also the
optimal dose regimens for treatment (acute bleeding episodes, prophylaxis, immune
tolerance induction, infant delivery, surgery and dental procedures) and a range of
management issues. To ensure the recruitment of adequate sample sizes to provide
statistical power, multi-centred controlled trials (both national and international studies)
are encouraged where possible to provide higher-level evidence to support clinical
practice guidelines.
National Registry
It would also be advantageous to Australians with bleeding disorders and for generating
research that is relevant to the Australian setting if an Australian Registry of patients with
bleeding disorders was established. This registry, which should include data from all the
states and territories of Australia, would be a valuable mechanism for reporting viral
transmission, adverse events and other complications of treatment, and could act as an
early warning system for detecting the presence of unknown immunogenic agents or
other possible contaminants in products. It could also provide useful data on long-term
outcomes of treatment options.
Immunosuppressive therapy
The evidence emerging from this systematic review indicates that immunosuppressive
therapy with cyclophosphamide is not recommended for patients with inhibitors, due to
poor haemostatic control and a high incidence of adverse events. In contrast, several
small case series have found that the immunosuppressant, rituximab, has a better short
term safety profile than those used previously, such as cyclophosphamide or
prednisolone. However, the body of evidence is insufficient to make recommendations
for its use. Further good quality well-designed research is needed to determine its safety
and effectiveness in both the short- and long-term for eliminating inhibitors.
Recombinant von Willebrand factor concentrates
There remains a theoretical risk of viral and prion transmission from the use of factor
concentrates that are derived from plasma. The development of a recombinant von
Willebrand factor concentrate should put patients at less risk of infectious diseases than
those containing plasma-derived products and support should be given to research this
potential therapeutic product.
140
Use of recombinant and plasma-derived Factor VIII and IX
Prophylaxis for von Willebrand disease patients
There have been several studies that reported beneficial effects of a DDAVP nasal spray
or a high dose of tranexamic acid as prophylaxis for menorrhagia in patients with von
Willebrand disease. However, sample sizes were generally too small to provide conclusive
evidence. Well-designed and controlled studies should be conducted to determine the
safety and effectiveness of different prophylaxis strategies, particularly with the less
invasive treatments, such as nasal sprays or mouthwashes that may be easily selfadministered by patients with menorrhagia. Similarly, further studies using a combination
of treatments, such as tranexamic acid and DDAVP are necessary to determine whether
they effectively reduce excessive menstrual bleeding in these patients.
Tolerisation
There is currently no consensus on the best method for tolerisation. Participation in
ongoing multi-centre studies on tolerisation for both haemophilia A and B patients (with
inhibitors) should be encouraged so that this expensive treatment option is used most
effectively. Moreover, comparative studies on the effectiveness and safety of high-dose
versus low-dose protocols for tolerisation are essential (e.g. the International Immune
Tolerance Study). More research is needed on treatment of long standing high titre
inhibitors or relapsed inhibitors. Controlled comparative studies are needed to confirm
findings from lower level evidence that the Malmö protocol is more effective in patients
with long standing inhibitors.
Surgery and dental procedures
Further study is required to assess the optimal duration of treatment after a surgical or
dental procedure. The current knowledge base regarding surgical procedures in patients
with blood clotting disorders consists solely of case series or case reports. Randomised
controlled trails are required to establish what protocol is safest and most effective.
Ascertaining response to rFVIIa
Activated rFVII has been found to be useful in patients with inhibitors, but futher
research is needed to ascertain the mechanisms of response from this treatment. No
evidence of a dose response relationship has been found (Dejgaard 2003).
Currently the response to treatment with rFVIIa is not able to be adequately measured.
New approaches to treatment
Although gene therapy for haemophilia A is still in its infancy, experimental studies
appear to show some promise and approximately five different trials, using vector
transgene expression, are in Phase I (Rick et al. 2003). The ultimate goal of gene therapy
is the long-term, sustained, therapeutic production of appropriate coagulant factors
without an immune response to the transgene product or vector. Alternative approaches
include: 1) the use of pre-messenger RNA repair, which use endogenous splicing
mechanisms to correct the defective RNA; 2) Gene-modified circulating endothelial
progenitor cells, which are capable of synthesising FVIII, may be isolated from
Use of recombinant and plasma-derived Factor VIII and IX
141
peripheral blood, expanded in culture, and modified to carry the normal FVIII gene; and
3) gene-modified stem cell therapy, which involves the use of multipotent adult
progenitor cells derived from marrow stroma. These stem cells may be genetically
modified to synthesise coagulation factors prior to retransplantation. However, it may
take several months before sufficient cells are generated for transplantation.
Determination of phenotype-genotype relationship
Further research should be done in determining the relationship between phenotypes and
genotypes in haemophilia and, in particular, in von Willebrand disease. While the
genomic sequence of von Willebrand factor gene is available, better techniques in
describing the phenotypes are needed for both clinical and research purposes. Through
collecting data on correlations between phenotypes and genotypes, it is hoped that better
classification of the disease may occur, which will allow more appropriate management
of the disorder.
Acquired Haemophilia
Research is needed to determine the safest and most effective immunosuppressant
therapy for use in acquired haemophilia.
142
Use of recombinant and plasma-derived Factor VIII and IX
Glossary and Abbreviations
ABDR
Australian Bleeding Disorders Registry
AHCDO
Australian Haemophilia Centre Directors’ Organisation
aPCCs
Activated prothrombin complex concentrates
AP-FVIII
Affinity-purified factor VIII
aPTT
Activated partial thromboplastin time
AHF-HP
Antihaemophilic factor—high purity
AHMAC
Australian Health Ministers’ Advisory Committee
AP-FVIII
Affinity-purified factor VIII
BDDrFVIII
B-domain deleted recombinant factor VIII
BT
Bleeding time—time for bleeding to stop from a superficial
incision
BU
Bethesda Units—a measure of inhibitor activity; the amount of
inhibitor that will inactivate 50% or 0.5 unit of a coagulation
factor during the incubation period
CI
Confidence interval
CJD
Creutzfeld-Jakob disease
CVAD
Central venous access device
Cytopaenia
Decrease in the number of blood cells
DDAVP
1-deamino-8-D-arginine vasopressin (desmopressin)
DIC
Disseminated intravascular coagulation
DVT
Deep vein thrombosis
ED
Days of exposure prior to inhibitor development
ELISA
Enzyme linked immunosorbent assay
Epistaxis
Nosebleed
Erythema
Redness of the skin
FEIBA
Factor Eight Inhibitor Bypassing Agent
FFP
Clinical fresh frozen plasma
Use of recombinant and plasma-derived Factor VIII and IX
143
FL-FVIII
Full-length FVIII
Fractionation
A process that uses heat to separate blood into its components
FII
Factor II, fibrinogen
FV
Factor V
FVII
Factor VII
FVIII:C
Factor VIII coagulant activity
FIX
Factor IX
FX
Factor X
FXI
Factor XI
FXIII
Factor XIII
HBV
Hepatitis B virus
HCV
Hepatitis C virus
Haemarthrosis
Bleeding into joint spaces
Haematoma
Bleeding into an organ or tissue
High responders
Exhibit high levels of inhibitors (>5 BU/ml) following factor
concentration administration
High titre
More than five Bethesda Units per millilitre (BU/ml)
HIV
Human immunodeficiency virus
Hyponatraemia
Abnormal decrease in blood sodium concentration
IgG
Immunoglobulin G
Inhibitor
An antibody that neutralises the function of factor concentrates
IQR
Inter quartile range
ITI
Immune Tolerance Induction
IU
International Units—internationally accepted unit of measurement
for amount of clotting factor
IVR
In vivo recovery
JBC
Jurisdictional Blood Committee
Low responders
Show minimal or no increase in inhibitor titres following factor
concentrate administration
144
Use of recombinant and plasma-derived Factor VIII and IX
Low titre
Less than or equal to five Bethesda Units per millilitre
Lymphopaenia
Decrease in lymphocytes in the blood
MASAC
Medical and Scientific Advisory Council (USA)
MI
Myocardial infarction
MTPs
Minimally treated patients—who received ≤ prior infusions
NBDR
National Bleeding Disorder Registry
Neutropaenia
Decrease in neutrophils in the blood
NHMRC
National Health and Medical Research Council
Pancytopaenia
Deficiency of all cell elements in the blood
Paraesthesia
Abnormal burning, prickling sensation
PBAC
Pictorial Blood Assessment Chart
PCCs
Prothrombin Complex Concentrates
PCR
Polymerase chain reaction
pdFIX
Plasma-derived factor IX
pdFVIII
Plasma-derived factor VIII
PFA-100
Platelet function analyser
PPH
Postpartum haemorrhage
PPT
Partial prothrombin time
Prophylaxis
An attempt to prevent bleeding episodes by administering
amounts of blood clotting factors
Pruritis
Itching of the skin
PT
Prothrombin time
PTPs
Previously treated patients
PUPs
Previously untreated patients
Rco
Ristocetin-cofactor activity
rFIX
Recombinant factor IX
rFVIIa
Activated recombinant factor VII
rFVIII
Recombinant factor VIII
Use of recombinant and plasma-derived Factor VIII and IX
145
RR
Relative risk/rate ratio
SD
Standard deviation
Tachyphylaxis
Rapidly decreasing response to a drug after administration of a
few doses
Tolerisation
Induction of immune tolerance (desensitisation to blood clotting
factors)
Teichopsia
Jagged visual sensation resembling the walls of a medieval town
Thrombophlebitis
Inflammation of a vein associated with thrombus formation
Urticaria
Transient allergic reaction of the skin—severe itching
vCJD
variant Creutzfeldt-Jakob disease
vWD
von Willebrand disease
vWF
von Willebrand factor
vWF:Ag
von Willebrand factor antigen
vWF:CB
von Willebrand factor-collagen binding
vWF:FVIIIB
von Willebrand factor-factor VIII binding
vWF:Rco
von Willebrand factor-ristocetin cofactor binding
146
Use of recombinant and plasma-derived Factor VIII and IX
References
AHMAC 2003, Report of the Working Party on the supply and use of factor VIII and factor IX in
Australia, Department of Health and Ageing, Canberra.
AIHW 2004, National Hospital Morbidity Database, viewed 25 June 2004, www.aihw.gov.au
Ananyeva, N, Khrenov, A et al. 2004, 'Treating haemophilia A with recombinant blood
factors: a comparison', Expert Opin Pharmacother, 5 (5), pp. 1061–1070.
Arkin, S, Rose, E et al. 1991, 'Clinical efficacy of recombinant factor VIII', Semin Hematol,
28 (2 Suppl. 1), pp. 47–51.
Association of Hemophilia Clinic Directors of Canada 1995a, 'Hemophilia and von
Willebrand's disease: 1. Diagnosis, comprehensive care and assessment. Association of
Hemophilia Clinic Directors of Canada. Updated 1999', Can Med Assoc J, 153 (1), pp. 19–
25.
Association of Hemophilia Clinic Directors of Canada 1995b, 'Hemophilia and von
Willebrand's disease: 2. Management. Association of Hemophilia Clinic Directors of
Canada', Can Med Assoc J, 153 (2), pp. 147–157.
Auerswald, G, Eberspacher, B et al. 2002, 'Successful treatment of patients with von
Willebrand disease using a high-purity double-virus inactivated factor VIII/von
Willebrand factor concentrate (Immunate (registered trademark))', Semin Thromb Hemost,
28 (2), pp. 203–213.
Australian Bleeding Disorder Registry 2004, ABDR Report, National Blood Authority,
Canberra.
Australian Haemophilia Centre Directors' Organisation 2004, Guidelines for the treatment of
inhibitors in Haemophilia A, Canberra.
Avanoglu, A, Celik, A et al. 1999, 'Safer circumcision in patients with haemophilia: The
use of fibrin glue for local haemostasis', BJU International, 83 (1), pp. 91–94.
Aygoren-Pursun, E, Scharrer, I et al. 1997, 'A multicenter pharmacosurveillance study for
the evaluation of the efficacy and safety of recombinant factor VIII in the treatment of
patients with hemophilia A', Thromb Haemost, 78 (5), pp. 1352–1356.
Bastounis, E, Pikoulis, E et al. 2000, 'General surgery in haemophiliac patients', Postgrad
Med J, 76 (898), pp. 494–495.
Baudo, F, De Cataldo, F & Gaidano, G 2004, 'Treatment of acquired factor VIII
inhibitor with recombinant activated factor VIIa: data from the Italian registry of
acquired hemophilia', Haematologica, 89 (6), pp. 759–761.
Berntorp, E 1994, 'Plasma product treatment in various types of von Willebrand's
disease', Haemostasis, 24 (5), pp. 289–297.
Berntorp, E 1997, 'Second generation, B-domain deleted recombinant factor VIII',
Thromb Haemost, 78 (1), pp. 256–260.
Use of recombinant and plasma-derived Factor VIII and IX
147
Berntorp, E 1999, 'von Willebrand factor containing factor VIII concentrates',
Haemophilia, 5 (Suppl. 2), pp. 60–63.
Berntorp, E, Astermark, J et al. 2003. 'Consensus perspective on prophylactic therapy for
haemophilia: Summary statement', Haemophilia, 9 (Suppl. 1), pp. 1–4.
Bjorkman, S, Shapiro, AD & Berntorp, E 2001, 'Pharmacokinetics of recombinant factor
IX in relation to age of the patient: implications for dosing in prophylaxis', Haemophilia, 7
(2), pp. 133–139.
Bolton-Maggs, PH 2000, 'Factor XI deficiency and its management', Haemophilia, 6
(Suppl 1), pp. 100–109.
Bolton-Maggs, PH & Pasi, KJ 2003, 'Haemophilias A and B', Lancet, 361 (9371), pp.
1801–1809.
Bolton-Maggs, PH, Perry, DJ et al. 2004, 'The rare coagulation disorders—review with
guidelines for management from the United Kingdom Haemophilia Centre Doctors'
Organisation', Haemophilia, 10 (5), pp. 593–628.
Braunwald, E, Fauci, AS et al. 2001, Harrison's (15th Edition) principles of internal medicine,
Vol. 1 .15 McGraw-Hill Medical Publishing Division, New York.
Bray, GL, Gomperts, ED et al. 1994, 'A multicenter study of recombinant factor VIII
(recombinate): safety, efficacy, and inhibitor risk in previously untreated patients with
hemophilia A. The Recombinate Study Group', Blood, 83 (9), pp. 2428–2435.
Canadian Hemophilia Society 2004, Factor II deficiency (Prothrombin deficiency), viewed
7 January 2005, www.hemophilia.ca/en/2.3.2.php.
Carcao, MD & Aledort, L 2004, 'Prophylactic factor replacement in hemophilia', Blood
Rev, 18 (2), pp. 101-113.
Cattaneo, M, Moia, M et al. 1989, 'DDAVP shortens the prolonged bleeding times of
patients with severe von Willebrand disease treated with cryoprecipitate. Evidence for a
mechanism of action independent of released von Willebrand factor', Blood, 74 (6),
pp. 1972–1975.
Chalmers, EA. 2004, 'Haemophilia and the newborn', Blood Rev, 18 (2), pp. 85–92.
Chediak, JR, Alban, GM & Maxey, B 1986, 'von Willebrand's disease and pregnancy:
management during delivery and outcome of offspring', Am J Obstet Gynecol, 155 (3),
pp. 618–624.
Chuansumrit, A, Hathirat, P et al. 1993, 'Response of patients with bleeding disorder to
DDAVP administration', Southeast Asian J Trop Med Public Health, 24 (Suppl 1), pp. 174–
179.
Clarke, M & Oxman, AD 2000, Cochrane reviewers' handbook 4.0 , 'The Cochrane
Collaboration', viewed 26 July 2004, www.cochrane.org/cochrane/hbook.htm.
Collins, P, Macartney, N et al. 2004, 'A population based, unselected, consecutive cohort
of patients with acquired haemophilia A', Br J Haematol, 124 (1), pp. 86–90.
148
Use of recombinant and plasma-derived Factor VIII and IX
Cooper, H & Hedges, LV (eds.) 1994), The handbook of research synthesis, Russel Sage
Foundation, New York.
Daffos, F, Forestier, F et al. 1988, 'Prenatal diagnosis and management of bleeding
disorders with fetal blood sampling', Am J Obstet Gynecol, 158 (4), pp. 939–946.
de la Fuente, B, Kasper, CK et al. 1985, 'Response of patients with mild and moderate
hemophilia A and von Willebrand's disease to treatment with desmopressin', Ann Intern
Med, 103 (1), pp. 6–14.
De Sio, L, Mariani, G & Muzzucconi, G 1985, 'Comparison between subcutaneous and
intravenous DDAVP in mild and moderate Hemophilia A', Thromb Haemost, 54 (2),
pp. 387–389.
Dejgaard, A 2003, 'Update on Novo Nordisk's clinical trial programme on NovoSeven®',
Blood Coag Fibrinolysis, 14 (Suppl 1), S39–S41.
Delgado, J, Jimenez-Yuste, V et al. 2003, 'Acquired haemophilia: review and metaanalysis focused on therapy and prognostic factors', Br J Haematol, 121 (1), pp. 21–35.
Delgado, J, Villar, A et al. 2002, 'Acquired hemophilia: a single-center survey with
emphasis on immunotherapy and treatment-related side-effects', Eur J Haematol, 69 (3),
pp. 158–164.
Department of Health and Ageing 2004, Access to recombinant factor VIII and IX, viewed 14
September 2004, www.nba.gov.au/current.htm.
Department of Health and Ageing 2003, Report of the Expert Advisory Group on Hepatitis C
and Plasma in 1990, Canberra.
Dewald, DL, Briggs, B & Smith, RE 1980, 'Intranasal administration of desmopressin
acetate (DDAVP) to hemophiliacs', Thromb Res, 18 (5), pp. 617–622.
Di Paola, J, Nugent, D & Young, G 2001, 'Current therapy for rare factor deficiencies',
Haemophilia, 7 (Suppl 1), pp. 16–22.
DiMichele, DM 1998, 'Immune tolerance: A synopsis of the international experience',
Haemophilia, 4 (4), pp. 568–573.
Djulbegovic, B, Marasa, M et al. 1996, 'Safety and efficacy of purified factor IX
concentrate and antifibrinolytic agents for dental extractions in hemophilia B', Am J
Hematol, 51 (2), pp. 168–170.
Dobrkovska, A, Krzensk, U & Chediak, JR 1998, 'Pharmacokinetics, efficacy and safety
of Humate-P in von Willebrand disease', Haemophilia, 4 (Suppl 3), pp. 33–39.
Downs, SH & Black, N 1998, 'The feasibility of creating a checklist for the assessment of
the methodological quality both of randomised and non-randomised studies of health
care interventions', J Epidemiol Community Health, 52 (6), pp. 377–384.
Dunn, AL, Powers, JR et al. 2000, 'Adverse events during use of intranasal desmopressin
acetate for haemophilia A and von Willebrand disease: a case report and review of 40
patients', Haemophilia, 6 (1), pp. 11–14.
Use of recombinant and plasma-derived Factor VIII and IX
149
Economides, DL, Kadir, RA & Lee, CA 1999, 'Inherited bleeding disorders in obstetrics
and gynaecology', Br J Obstet Gynaecol, 106 (1), pp. 5–13.
Ewenstein, BM, Gomperts, ED et al. 2004, 'Inhibitor development in patients receiving
recombinant factor VIII (Recombinate rAHF/Bioclate): a prospective pharmacovigilance
study', Haemophilia, 10 (5), pp. 491–498.
Ewenstein, BM, Joist, JH et al. 2002, 'Pharmacokinetic analysis of plasma-derived and
recombinant F IX concentrates in previously treated patients with moderate or severe
hemophilia B', Transfusion, 42 (2), pp. 190–197.
Favaloro, E, Lloyd, J et al. 2005, 'A cross-over, multi-centre study to compare the
pharmacokinetics of two factor concentrates (Biostate and AHF (High Purity)) in people
with von Willebrand disorder (Draft).' Manuscript in preparation.
Federici, AB, Baudo, F et al. 2002, 'Clinical efficacy of highly purified, doubly virusinactivated factor VIII/von Willebrand factor concentrate (Fanhdi) in the treatment of
von Willebrand disease: a retrospective clinical study', Haemophilia, 8 (6), pp. 761–767.
Federici, AB, Mazurier, C et al. 2004, 'Biologic response to desmopressin in patients with
severe type 1 and type 2 von Willebrand disease: results of a multicenter European study',
Blood, 103 (6), pp. 2032–2038.
Federici, AB, Sacco, R et al. 2000, 'Optimising local therapy during oral surgery in
patients with von Willebrand disease: effective results from a retrospective analysis of 63
cases', Haemophilia, 6 (2), pp. 71–77.
Fijnvandraat, K, Berntorp, E et al. 1997, 'Recombinant, B-domain deleted factor VIII (rVIII SQ): pharmacokinetics and initial safety aspects in hemophilia A patients', Thromb
Haemost, 77 (2), pp. 298–302.
Franchini, M, Gandini, G et al. 2002, 'Evaluation of the PFA-100 system for monitoring
desmopressin therapy in patients with type 1 von Willebrand's disease', Haematologica, 87
(6), p. 670.
Franchini, M, Rossetti, G et al. 2003, 'Efficacy and safety of factor VIII/von Willebrand's
factor concentrate (Haemate-P) in preventing bleeding during surgery or invasive
procedures in patients with von Willebrand disease', Haematologica, 88 (11), pp. 1279–
1283.
Ghirardini, A, Chistolini, A et al. 1988,'Clinical evaluation of subcutaneously
administered DDAVP', Thromb Res, 49 (3), pp. 363–372.
Ghirardini, A, Mariani, G et al. 1987, 'Concentrated DDAVP: further improvement in
the management of mild factor VIII deficiencies', Thromb Haemost, 58 (3), pp. 896–898.
Giangrande, DP 1996, Inhibitory antibodies, UK Haemophilia Centre Directors'
Organisation, viewed 6 July 2004, ww.medicine.ox.ac.uk/ohc/Inhibi.htm.
Giangrande, PL 2002, 'Safety and efficacy of KOGENATEI Bayer in previously
untreated patients (PUPs) and minimally treated patients (MTPs)', Haemophilia, 8 (Suppl
2), pp. 19–22.
150
Use of recombinant and plasma-derived Factor VIII and IX
Giangrande, PLF 1998, 'Management of pregnancy in carriers of haemophilia',
Haemophilia, 4 (6), 779–784.
Giles, AR, Rivard, GE et al. 1998, 'Surveillance for factor VIII inhibitor development in
the Canadian Hemophilia A population following the widespread introduction of
recombinant factor VIII replacement therapy', Transfus Sci, 19 (2), pp. 139–148.
Gill, JC, Ewenstein, BM et al. 2003, 'Successful treatment of urgent bleeding in von
Willebrand disease with factor VIII/VWF concentrate (Humate-P): use of the ristocetin
cofactor assay (VWF:RCo) to measure potency and to guide therapy', Haemophilia, 9 (6),
pp. 688–695.
Gill, JC, Ottum, M & Schwartz, B 2002, 'Evaluation of high concentration intranasal and
intravenous desmopressin in pediatric patients with mild hemophilia A or mild-tomoderate type 1 von Willebrand disease', J Pediatr, 140 (5), pp. 595–599.
Greer, IA, Lowe, GDO et al. 1991, 'Haemorrhagic problems in obstetrics and
gynaecology in patients with congenital coagulopathies', Br J Obstet Gynaecol, 98 (9),
pp. 909–918.
Gringeri, A 1998, 'Evaluation study on prophylaxis: a randomised Italian trial (ESPRIT).
One year report [abstract]', Haemophilia, 4 (3), p. 171.
Gringeri, A 2003, 'Prospective controlled studies on prophylaxis: an Italian approach',
Haemophilia, 9 (Suppl 1), pp. 38–42; discussion p. 43.
Gringeri, A, Tagliaferri, A et al. 2004, 'Efficacy and inhibitor development in previously
treated patients with haemophilia A switched to a B domain-deleted recombinant factor
VIII', Br J Haematol, 126 (3), pp. 398–404.
Gruppo, R, Brown, D et al. 2004, 'Increased breakthrough bleeding during prophylaxis
with B-domain deleted factor VIII - a robust meta-analytic finding', Haemophilia, 10,
pp. 1–3.
Gruppo, R, Brown, D et al. 2003, 'Comparative effectiveness of full-length and Bdomain deleted factor VIII for prophylaxis - a meta-analysis', Haemophilia, 9 (3), pp. 251–
260.
Haemophilia Foundation Australia 2004a, Do we have to wait for another disaster like Hepatitis
C before governments will act?, viewed 7 July 2004,
www.haemophilia.org.au/knowledge/files/143/june172004final.pdf.
Haemophilia Foundation Australia 2004b, Haemophilia Foundation Australia, viewed 4
August 2004, www.haemophilia.org.au.
Haemophilia Foundation Australia Medical Advisory Panel 2000, Guidelines on therapeutic
products to treat haemophilia and other hereditary coagulation disorders in Australia, viewed 26 July
2004, www.ahcdo.org.au/sitebuilder/guidelines/files/3/guidelinesmarch00.pdf.
Hay, C, Mauser-Bunschoten, E et al. 2000a, Protocol: An International randomised controlled
trial of immune-tolerance induction, Manchester.
Use of recombinant and plasma-derived Factor VIII and IX
151
Hay, CR, Baglin, TP et al. 2000b, 'The diagnosis and management of factor VIII and IX
inhibitors: a guideline from the UK Haemophilia Centre Doctors' Organization
(UKHCDO)', Br J Haematol, 111 (1), pp. 78–90.
Hitchens, K 2004, 'Special Report: Helping Hemophiliacs', Drug Topics, 148 (HSE17).
Hoyer, LW, Carta, CA et al. 1985, 'Prenatal diagnosis of classic hemophilia (hemophilia
A) by immunoradiometric assays', Blood, 65 (6), pp. 1312–1317.
Israels, S 2004a, Factor VII deficiency, eMedicine, viewed 16 December 2004,
www.emedicine.com/ped/topic3041.htm.
Israels, S 2004b, Factor XIII deficiency, eMedicine, viewed 16 December 2004,
www.emedicine.com/ped/topic3040.htm.
JBC FVIII/FIX working party & National Blood Authority 2004, Development of evidencebased clinical practice guidelines for the use of recombinant and plasma-derived factor VIII and IX
(meeting), Melbourne, 15 December 2004.
Kadir, RA, Economides, DL et al. 1997, 'The obstetric experience of carriers of
haemophilia', Br J Obstet Gynaecol, 104 (7), pp. 803–810.
Kadir, RA., Lee, CA et al. 1998, 'Pregnancy in women with von Willebrand's disease or
factor XI deficiency', Br J Obstet Gynaecol, 105 (3), pp. 314–321.
Kadir, RA., Lee, CA et al. 2002, 'DDAVP nasal spray for treatment of menorrhagia in
women with inherited bleeding disorders: a randomized placebo-controlled crossover
study', Haemophilia, 8 (6), pp. 787–793.
Kasper, CK 2004, Hereditary plasma clotting factor disorders and their management. Treatment of
Hemophilia Monograph Series, No. 4, World Federation of Hemophilia, Montreal, Canada.
Kelly, KM, Butler, RB et al. 1997, 'Superior in vivo response of recombinant factor VIII
concentrate in children with hemophilia A', J Pediatr, 130 (4), pp. 537–540.
Khan, KS, Ter Riet, G et al. 2001, Undertaking systematic reviews of research on effectiveness.
CRD's guidance for those carrying out or commissioning reviews, NHS Centre for Reviews and
Dissemination, University of York, York.
Kingdon, HS & Lundblad, RL 2002, 'An adventure in biotechnology: the development of
haemophilia A therapeutics - from whole-blood transfusion to recombinant DNA to
gene therapy', Biotechnol Appl Biochem, 35 (Pt 2), pp. 141–148.
Kisker, CT, Eisberg, A & Schwartz, B 2003, 'Prophylaxis in factor IX deficiency product
and patient variation', Haemophilia, 9 (3), pp. 279–284.
Kreuz, W, Ettingshausen, CE et al. 2002, 'Inhibitor development in previously untreated
patients with hemophilia A: a prospective long-term follow-up comparing plasmaderived and recombinant products', Semin Thromb Hemost, 28 (3), pp. 285–290.
Kulkarni, R, Aledort, LM et al. 2001, 'Therapeutic choices for patients with hemophilia
and high-titer inhibitors', Am J Hematol, 67 (4), pp. 240–246.
152
Use of recombinant and plasma-derived Factor VIII and IX
Kulkarni, R & Lusher, J 2001, 'Perinatal management of newborns with haemophilia', Br
J Haematol, 112 (2), pp. 264–274.
Laffan, M, Brown, SA et al. 2004, 'The diagnosis of von Willebrand disease: a guideline
from the UK Haemophilia Centre Doctors' Organization', Haemophilia, 10 (3), pp. 199–
217.
Leissinger, C, Becton, D et al. 2001, 'High-dose DDAVP intranasal spray (Stimate) for
the prevention and treatment of bleeding in patients with mild haemophilia A, mild or
moderate type 1 von Willebrand disease and symptomatic carriers of haemophilia A',
Haemophilia, 7 (3), pp. 258–266.
Lillicrap, D, Poon, MC et al. 2002, 'Efficacy and safety of the factor VIII/von Willebrand
factor concentrate, haemate-P/humate-P: ristocetin cofactor unit dosing in patients with
von Willebrand disease', Thromb Haemost, 87 (2), pp. 224–230.
Ljung, R 2004, 'Central venous catheters in children with haemophilia', Blood Rev, 18 (2),
pp. 93–100.
Ljung, R, Lindgren, AC et al. 1994, 'Normal vaginal delivery is to be recommended for
haemophilia carrier gravidae', Acta Paediatr, 83 (6), pp. 609–611.
Lloyd Jones, M, Wight, J et al. 2003, 'Control of bleeding in patients with haemophilia A
with inhibitors: a systematic review', Haemophilia, 9 (4), pp. 464–520.
Lorenzo, JI, Lopez, A et al. 2001, 'Incidence of factor VIII inhibitors in severe
haemophilia: the importance of patient age', Br J Haematol, 113 (3), pp. 600–603.
Lusher, J, Abildgaard, C et al. 2004, 'Human recombinant DNA-derived antihemophilic
factor in the treatment of previously untreated patients with hemophilia A: final report
on a hallmark clinical investigation', J Thromb Haemost, 2 (4), pp. 574–583.
Lusher, JM, Arkin, S et al. 1993, 'Recombinant factor VIII for the treatment of
previously untreated patients with hemophilia A—Safety, efficacy, and development of
inhibitors', N Engl J Med, 328 (7), pp. 453–459.
Manco-Johnson, MJ, Nuss, R et al. 2000, 'Joint evaluation instruments for children and
adults with haemophilia', Haemophilia, 6 (6), pp. 649–657.
Manco-Johnson, MJ & Blanchette, VS 2003, 'North American prophylaxis studies for
persons with severe haemophilia: Background, rationale and design', Haemophilia, 9
(SUPPL. 1), pp. 44–49.
Manno, CS 1999, 'Treatment options for bleeding episodes in patients undergoing
immune tolerance therapy', Haemophilia, 5 (Suppl 3), pp. 33–41.
Mannucci, PM 2001, 'How I treat patients with von Willebrand disease', Blood, 97 (7),
pp. 1915–1919.
Mannucci, PM, Bettega, D & Cattaneo, M 1992, 'Patterns of development of
tachyphylaxis in patients with haemophilia and von Willebrand disease after repeated
doses of desmopressin (DDAVP)', Br J Haematol, 82 (1), pp. 87–93.
Use of recombinant and plasma-derived Factor VIII and IX
153
Mannucci, PM, Brettler, DB et al. 1994, 'Immune status of human immunodeficiency
virus seropositive and seronegative hemophiliacs infused for 3.5 years with recombinant
factor VIII. The Kogenate Study Group', Blood, 83 (7), pp. 1958–1962.
Mannucci, PM, Chediak, J et al. 2002, 'Treatment of von Willebrand disease with a highpurity factor VIII/von Willebrand factor concentrate: a prospective, multicenter study',
Blood, 99 (2), pp. 450–456.
Mannucci, PM, Vicente, V et al. 1987, 'Intravenous and subcutaneous administration of
desmopressin (DDAVP) to hemophiliacs: pharmacokinetics and factor VIII responses',
Thromb Haemost, 58 (4), pp. 1037–1039.
Mariana, G, Ciavarella, N & Mazzucconi, MG 1984, 'Evaluation of the effectiveness of
DDAVP in surgery and in bleeding episodes in haemophilia and von Willebrand's
disease. A study on 43 patients', Clin Lab Haematol, 6 (3), pp. 229–238.
Medical and Scientific Advisory Council (MASAC) 2001, Recommendation concerning
prophylaxis (Prophylactic administration of clotting factor concentration to prevent bleeding), National
Hemophilia Foundation, New York.
Mohri, H 2002, 'High dose of tranexamic acid for treatment of severe menorrhagia in
patients with von Willebrand disease', J Thromb Thrombolysis, 14 (3), pp. 255–257.
Morfini, M, Longo, G et al. 1992, 'Pharmacokinetic properties of recombinant factor
VIII compared with a monoclonally purified concentrate (Hemofil M). The Recombinate
Study Group', Thromb Haemost, 68 (4), pp. 433–435.
Morrison, AE, Ludlam, CA & Kessler, C 1993, 'Use of porcine factor VIII in the
treatment of patients with acquired hemophilia', Blood, 81 (6), pp. 1513–1520.
Mulrow, CD, Cook, DJ & Davidoff, F 1997, 'Systematic reviews: Critical links in the
great chain of evidence', Ann Intern Med, (126), pp. 389–391.
NHMRC 2000a, How to review the evidence: systematic identification and review of the scientific
literature, National Health and Medical Research Council, Canberra.
NHMRC 2000b, How to use the evidence: assessment and application of scientific evidence, National
Health and Medical Research Council, Canberra.
NHMRC Guidelines Assessment Register Consultants Working Party 2004, Minimum
requirements for evidence tables, Guideline Assessment Register, National Health and Medical
Research Council, Canberra.
Nilsson, IM., Berntorp, E et al. 1992, 'Twenty-five years' experience of prophylactic
treatment in severe haemophilia A and B', J Intern Med, 232 (1), pp. 25–32.
Nilsson, IM & Hedner, U 1976, 'Immunosuppressive treatment in haemophiliacs with
inhibitors to factor VIII and factor IX', Scand J Haematol, 16 (5), pp. 369–382.
Nitu-Whalley, IC, Griffioen, A et al. 2001, 'Retrospective review of the management of
elective surgery with desmopressin and clotting factor concentrates in patients with von
Willebrand disease', Am J Hematol, 66 (4), pp. 280–284.
154
Use of recombinant and plasma-derived Factor VIII and IX
Nolan, B, White, B et al. 2000, 'Desmopressin: therapeutic limitations in children and
adults with inherited coagulation disorders', Br J Haematol, 109 (4), pp. 865–869.
Ong, YL., Hull, DR & Mayne, EE 1998, 'Menorrhagia in von Willebrand disease
successfully treated with single daily dose tranexamic acid', Haemophilia, 4 (1), pp. 63–65.
Paisley, S, Wight, J et al. 2003, 'The management of inhibitors in haemophilia A:
introduction and systematic review of current practice', Haemophilia, 9 (4), pp. 405–417.
Pall Corporation 2004, Pall Corp prion removal technology featured at International Transmissible
Spongiform Encephalopathy conference (press release), viewed 6 January 2005,
www.pall.com/news_articles_34853.asp.
Palmer, BF, Gates, JR & Lader, M 2003, 'Causes and management of hyponatremia', Ann
Pharmacother, 37 (11), pp. 1694–1702.
Panicker, J, Warrier, I et al. 2003, 'The overall effectiveness of prophylaxis in severe
haemophilia', Haemophilia, 9 (3), pp. 272–278.
Petrini, P 2001, 'What factors should influence the dosage and interval of prophylactic
treatment in patients with severe haemophilia A and B?' Haemophilia, 7 (1), pp. 99–102.
Pettersson, H, Nilsson, IM et al. 1981, 'Radiologic evaluation of prophylaxis in severe
haemophilia', Acta Paediatr Scand, 70 (4), pp. 565–570.
Pollmann, H, Richter, H et al. 1999, 'When are children diagnosed as having severe
haemophilia and when do they start to bleed? A 10-year single-centre PUP study', Eur J
Pediatr, 158 (Suppl 3), S166–170.
Poon, MC, Lillicrap, D et al. 2002, 'Recombinant factor IX recovery and inhibitor safety:
A Canadian post-licensure surveillance study', Thromb Haemost, 87 (3), pp. 431–435.
Powell, JS, Ragni, MV et al. 2003, 'Phase 1 trial of FVIII gene transfer for severe
hemophilia A using a retroviral construct administered by peripheral intravenous
infusion', Blood, 102 (6), pp. 2038–2045.
Ragni, MV, Pasi, KJ et al. 2002, 'Use of recombinant factor IX in subjects with
haemophilia B undergoing surgery', Haemophilia, 8 (2), pp. 91–97.
Rakocz, M, Mazar, A et al. 1993, 'Dental extractions in patients with bleeding disorders.
The use of fibrin glue', Oral Surg Oral Med Oral Pathol, 75 (3), pp. 280–282.
Rick, ME, Walsh, CE & Key, NS 2003, 'Congenital bleeding disorders', Hematology (Am
Soc Hematol Educ Program), pp. 559–574.
Rodeghiero, F, Castaman, G & Mannucci, PM 1996, 'Prospective multicenter study on
subcutaneous concentrated desmopressin for home treatment of patients with von
Willebrand disease and mild or moderate hemophilia A', Thromb Haemost, 76 (5), pp. 692–
696.
Rodriguez-Merchan, EC, Wiedel, JD et al. 2003, 'Elective orthopaedic surgery for
inhibitor patients', Haemophilia, 9 (5), pp. 625–631.
Use of recombinant and plasma-derived Factor VIII and IX
155
Rose, EH & Aledort, LM 1991, 'Nasal spray desmopressin (DDAVP) for mild
hemophilia A and von Willebrand disease', Ann Intern Med, 114 (7), pp. 563–568.
Roth, DA, Kessler, CM et al. 2001a, 'Human recombinant factor IX: safety and efficacy
studies in hemophilia B patients previously treated with plasma-derived factor IX
concentrates', Blood, 98 (13), pp. 3600–3606.
Roth, DA, Tawa, NE et al. 2001b, 'Nonviral transfer of the gene encoding coagulation
factor VIII in patients with severe hemophilia A', N Engl J Med, 344 (23), pp. 1735–1742.
Rothschild, C, Laurian, Y et al. 1998, 'French previously untreated patients with severe
hemophilia A after exposure to recombinant factor VIII: incidence of inhibitor and
evaluation of immune tolerance', Thromb Haemost, 80 (5), pp. 779–783.
Sallah, S 2004, 'Treatment of acquired haemophilia with factor eight inhibitor bypassing
activity', Haemophilia, 10 (2), pp. 169–173.
Sallah, S & Wan, JY 2003, 'Efficacy of 2-chlorodeoxyadenosine in refractory factor VIII
inhibitors in persons without hemophilia', Blood, 101 (3), pp. 943–945.
Santagostino, E, Gringeri, A et al. 2003, 'Long-term safety and feasibility of arteriovenous
fistulae as vascular accesses in children with haemophilia: a prospective study', Br J
Haematol, 123 (3), pp. 502–506.
Saulnier, J, Marey, A et al. 1994, 'Evaluation of desmopressin for dental extractions in
patients with hemostatic disorders.' Oral Surg Oral Med Oral Pathol, 77 (1), pp. 6–12.
Scharrer, I 1995, 'The need for highly purified products to treat hemophilia B', Acta
Haematol, 94 (Suppl 1), pp. 2–7.
Scharrer, I 2002, 'Experience with KOGENATEI Bayer in surgical procedures',
Haemophilia, 8 (Suppl 2), pp. 15–18.
Scharrer, I, Brackmann, HH et al. 2000, 'Efficacy of a sucrose-formulated recombinant
factor VIII used for 22 surgical procedures in patients with severe haemophilia A',
Haemophilia, 6 (6), pp. 614–618.
Schiavoni, M, Ettore, C et al. 1983, 'Tranexamic acid as prophylaxis in haemophilia. A
randomized double-blind study [abstract]', Thromb Haemost, pp. 50, 444.
Schulman, S, Johnsson, H & Lindstedt, M 1991, 'Desmopressin as a haemostatic agent in
patients with disorders of primary haemostasis', Acta Chir Eur J Surg, 157 (11-12),
pp. 647–652.
Schwartz, RS, Abildgaard, CF et al. 1990, 'Human recombinant DNA-derived
antihemophilic factor (factor VIII) in the treatment of hemophilia A', N Engl J Med, 323
(26), pp. 1800–1805.
Senate Community Affairs References Committee 2004, Hepatitis C and the blood supply in
Australia, Senate Printing Unit, Parliament House, Canberra.
156
Use of recombinant and plasma-derived Factor VIII and IX
Seremetis, S, Lusher, JM et al. 1999, 'Human recombinant DNA-derived antihaemophilic
factor (factor VIII) in the treatment of haemophilia A: conclusions of a 5-year study of
home therapy. The KOGENATE Study Group', Haemophilia, 5 (1), pp. 9–16.
Siegel, J 2004, Factor XI deficiency, eMedicine, viewed 16 December 2004,
www.emedicine.com/med/topic3515.htm.
Sindet-Pedersen, S, Stenbjerg, S & Ingerslev, J 1988, 'Control of gingival hemorrhage in
hemophilic patients by inhibition of fibrinolysis with tranexamic acid', J Periodontal Res, 23
(1), pp. 72–74.
Soucie, JM, Nuss, R et al. 2000, 'Mortality among males with hemophilia: relations with
source of medical care', Blood, 96 (2), pp. 437–442.
Soucie, JM, Symons, J et al. 2001, 'Home-based factor infusion therapy and
hospitalization for bleeding complications among males with haemophilia', Haemophilia, 7
(2), pp. 198–206.
Soucie, JM, Siwak, EB et al. 2004, 'Human parvovirus B19 in young male patients with
hemophilia A: associations with treatment product exposure and joint range-of-motion
limitation', Transfusion, 44 (8), pp. 1179–1185.
Stajcic, Z, Baklaja, R et al. 1989, 'Primary wound closure in haemophiliacs undergoing
dental extractions', Int J Oral Maxillofac Surg, 18 (1), pp. 14–16.
Stasi, R, Brunetti, M et al. 2004, 'Selective B-cell depletion with rituximab for the
treatment of patients with acquired hemophilia', Blood, 103 (12), 4424–4428.
Stata Corporation 2004, Intercooled Stata 8.2 for Windows [computer program], Stata
Corporation, College City, Texas.
Steen, CJ & Schwartz, RA 2004, Factor X, eMedicine, viewed 16 December 2004,
www.emedicine.com/med/topic3495.htm.
Stieltjes, N, Altisent, C et al. 2004, 'Continuous infusion of B-domain deleted
recombinant factor VIII (ReFacto) in patients with haemophilia A undergoing surgery:
clinical experience', Haemophilia, 10 (5), pp. 452–458.
Street, AM & Ekert, H 1996, 'Haemophilia—darkest hours before the dawn', Med J Aust,
164 (8), pp. 453–455.
Stubbs, M & Lloyd, J 2001, 'A protocol for the dental management of von Willebrand's
disease, haemophilia A and haemophilia B', Aust Dent J, 46 (1), pp. 37–40.
Tarantino, MD, Collins, PW et al. 2004, 'Clinical evaluation of an advanced category
antihaemophilic factor prepared using a plasma/albumin-free method: pharmacokinetics,
efficacy, and safety in previously treated patients with haemophilia A', Haemophilia, 10 (5),
pp. 428–437.
Thompson, AR, Gill, JC et al. 2004, 'Successful treatment for patients with von
Willebrand disease undergoing urgent surgery using factor VIII/VWF concentrate
(Humate-P)', Haemophilia, 10 (1), pp. 42–51.
Use of recombinant and plasma-derived Factor VIII and IX
157
United Kingdom Haemophilia Centre Doctors' Organisation 2003, 'Guidelines on the
selection and use of therapeutic products to treat haemophilia and other hereditary
bleeding disorders', Haemophilia, 9 (1), pp. 1–23.
Valentino, LA, Ewenstein, B et al. 2004, 'Central venous access devices in haemophilia',
Haemophilia, 10 (2), pp. 134–146.
van der Bom, JG, Mauser-Bunschoten, EP et al. 2003, 'Age at first treatment and
immune tolerance to factor VIII in severe hemophilia', Thromb Haemost, 89 (3), pp. 475–
479.
Verbruggen, B, Novakova, I et al. 1995, 'The Nijmegen modification of the Bethesda
assay for factor VIII:C inhibitors: improved specificity and reliability', Thromb Haemost, 73
(2), pp. 247–251.
Warrier, AI & Lusher, JM 1983, 'DDAVP: a useful alternative to blood components in
moderate hemophilia A and von Willebrand disease', J Pediatr, 102 (2), pp. 228–233.
Waymouth Resources Limited 2003, NSX Announcement: Proposed transaction with Living
Cell Technologies Ltd, viewed 12 July 2004,
www.newsx.com.au/ftp/news/WAY15102003175752.pdf.
White, G, Shapiro, A et al. 1998, 'Clinical evaluation of recombinant factor IX', Semin
Hematol, 35 (2 Suppl 2), pp. 33–38.
White, GC2nd, Courter, S et al. 1997, 'A multicenter study of recombinant factor VIII
(Recombinate) in previously treated patients with hemophilia A. The Recombinate
Previously Treated Patient Study Group', Thromb Haemost, 77 (4), pp. 660–667.
Wiestner, A, Cho, HJ et al. 2002, 'Rituximab in the treatment of acquired factor VIII
inhibitors', Blood, 100 (9), pp. 3426–3428.
Wight, J, Paisley, S & Knight, C 2003, 'Immune tolerance induction in patients with
haemophilia A with inhibitors: a systematic review', Haemophilia, 9 (4), pp. 436–463.
World Health Organization 2002, Delivery of Treatment of Haemophilia, Report of a Joint
WHO/WFH/ISTH Meeting, London, UK.
Yoshioka, A, Fukutake, K et al. 2003, 'Clinical evaluation of a recombinant factor VIII
preparation (Kogenate) in previously untreated patients with hemophilia A', Int J Hematol,
78 (5), pp. 467–474.
Yoshioka, A, Shima, M et al. 2001, 'Safety and efficacy of a new recombinant FVIII
formulated with sucrose (rFVIII-FS) in patients with haemophilia A: a long-term,
multicentre clinical study in Japan', Haemophilia, 7 (3), pp. 242–249.
Young, J, Hyde, C et al. 1999, 'Lung volume reduction surgery (LVRS) for chronic
obstructive pulmonary disease (COPD) with underlying severe emphysema.' Thorax, 54
(9), pp. 779–789.
Zanon, E, Martinelli, F et al. 2000, 'Proposal of a standard approach to dental extraction
in haemophilia patients. A case-control study with good results', Haemophilia, 6 (5),
pp. 533–536.
158
Use of recombinant and plasma-derived Factor VIII and IX
Ziv, O & Ragni, MV 2004, 'Bleeding manifestations in males with von Willebrand
disease', Haemophilia, 10 (2), pp. 162–168.
Use of recombinant and plasma-derived Factor VIII and IX
159
Appendix A
Terms of Reference for
Undertaking Guidelines
A. Services
1.
Develop national clinical guidelines for the use of recombinant factors VIII and IX
for patients with haemophilia A and B
2.
Develop national clinical guidelines for the use of plasma-derived factors VIII and
IX for patients with haemophilia A, B and von Willebrand disease
3.
Develop national tolerisation protocols for tasks 1 and 2 above
4.
Collect and consolidate information on product usage for patients with other rare
bleeding disorders, including an analysis on tolerisation within this group
5.
Attend and participate in education and implementation workshop(s)
B. Required contract material
The contractor shall produce both clinical practice guidelines and a final report to the
National Blood Authority.
1.
The guidelines shall include:
a.
b.
c.
d.
e.
Management of haemophilia A, B and von Willebrand disease
Adjunctive haemostatic agents
Treatment of bleeding episodes, including surgery, dental and prophylaxis
Delivery of infants
Complications of treatment, including:
(i) Inhibitor development
(ii) Infectious diseases and other safety and quality issues
f. Validation—data requirements for refined data analysis (clinical audit and
outcomes)
g. Other aspects of management
h. Selection of products
2.
160
In addition to a reconciliation of costs, the final report to the NBA shall identify
strategies and approaches for education of healthcare professionals and recipients
and identify potential research directions.
Use of recombinant and plasma-derived Factor VIII and IX
Appendix B
Working Party Membership
Contractors
Researchers:
Consultant
Methodologist:
Consultant Clinician:
Dr Petra Bywood, Research Officer
Ms Skye Newton, Research Officer
Ms Linda Mundy, Research Officer
Ms Tracy Merlin, Manager
Professor Janet Hiller, Director
Adelaide Health Technology Assessment
Department of Public Health, University of Adelaide
Dr Ben Saxon, Transfusion Medicine Specialist
Australian Red Cross Blood Service-Discovery
National Blood Authority
John Haines
National Blood Authority
Julianne Brettargh
National Blood Authority
Ray Carty
Scientific Officer, Corporate Services and Quality, National
Blood Authority
Ms Tamsen Marriott
Corporate Services and Quality, National Blood Authority
Ms Stephanie Gunn
Branch Manager Policy, Planning and Corporate Services,
National Blood Authority
Use of recombinant and plasma-derived Factor VIII and IX
161
Recombinant FVIII and FIX Working Party Members
Dr Chris Brook
Director, Rural and Regional Health and Aged Care Services
Victorian Department of Human Services
Ms Sharon Caris
Executive Director, Haemophilia Foundation Australia
Prof Brendon Kearney
(Chair)
Executive Director Clinical Systems, SA Department of
Human Services
Dr Ahti Lammi
Director of Haematology, New Childrens Hospital,
Westmead
Dr John Rowell
Director, Queensland Haemophilia Centre
Prof Alison Street
Director and Associate Professor of Medicine, Haematology
Department, Alfred Hospital, Melbourne
162
Use of recombinant and plasma-derived Factor VIII and IX
Appendix C
Australian haemophilia
treatment centres
Director
Address
A/Prof Ross Baker
Thrombosis and Haemophilia Service, Kirkman
House, Royal Perth Hospital, Wellington Street,
Perth, WA, 6000
Ph: 08 9224 2897/2667, Fax: 08 9224 8022,
[email protected]
Dr Chris Barnes
Department of Haematology/Oncology, Royal
Children’s Hospital, Flemington Road, Parkville,
VIC, 3052
Ph: 03 9345 5522, Fax: 03 9345 5099,
[email protected]
Dr David Jupe
Haematology Department, Royal Hobart Hospital,
GPO Box 1061L, Hobart, TAS, 7001
Ph: 03 6238 8418, Fax: 03 6263 7463,
[email protected]
Dr Ahti Lammi
Haematology Department, New Children’s
Hospital, Hawkesbury Road, Westmead, NSW,
2145
Ph: 02 9845 0000, Fax: 02 9845 3332,
[email protected]
Dr Michael Leahy
Haematology Department, Fremantle Hospital, PO
Box 480, Fremantle, WA, 6959
Ph: 08 9431 2886, Fax: 08 9431 2881,
[email protected]
A/Prof John Lloyd
Division of Haematology, IMVS, PO Box 14,
Rundle Mall, Adelaide, SA, 5000
Ph: 08 8222 5410, Fax: 8222 5963,
[email protected]
Dr Liane Lockwood
Haemophilia Centre, Royal Children’s Hospital,
Herston Road, Herston, QLD, 4029
Ph: 07 3636 9030, Fax: 07 3636 1552,
[email protected]
Dr Michael Pidcock
Department of Haematology, The Canberra
Hospital, Yamba Drive, Garran, ACT, 2606
Ph: 02 6244 2929/2894, Fax: 02 6244 2892,
[email protected]
Dr Jamie Price
Haematology Department, Princess Margaret
Hospital, GPO Box D184, West Perth, WA, 6005
Ph: 08 9340 8231, Fax: 08 9340 8384,
[email protected]
Dr Scott Dunkley
Haemophilia Treatment Centre, Royal Prince
Alfred Hospital, Missenden Road, Camperdown,
NSW, 2050
Ph: 02 9515 7013, Fax: 02 9515 8946
[email protected]
[email protected]
Dr John Rowell
Haematology Department, Royal Brisbane
Hospital, Herston Road, Herston, QLD, 4029
Ph: 07 3636 8067, Fax: 07 3252 1342,
[email protected]
Dr Susan Russell
Centre for Children’s Cancer & Blood Disorders,
Sydney Children’s Hospital, High Street, Randwick,
NSW, 2031
Ph: 02 9382 1690, Fax: 02 9382 1789,
[email protected]
Dr Ben Saxon
Department of Haematology, Women’s and
Children’s Hospital, 71 King William Road, North
Adelaide, SA, 5006
Ph: 08 8161 7327, Fax: 08 8161 6567,
[email protected]
Dr Michael Seldon
Haematology Department, Newcatle Mater
Misericordiae Hospital, Locked Bag 7, Hunter
Region Mail Centre, NSW, 2310
Ph: 02 4921 1216, Fax: 02 4960 2136,
[email protected]
Dr Sid Selva
Darwin Private Hospital, PO Box 42571,
Casuarina, NT, 0810
Ph: 08 8920 6176, Fax: 08 8920 6183,
[email protected]
Prof Alison Street
Department of Haematology, The Alfred Hospital,
Commercial Road, Prahran, VIC, 3181
Ph: 03 9276 3120, Fax: 03 9276 3781,
[email protected]
Use of recombinant and plasma-derived Factor VIII and IX
163
Appendix D Currently available product
information
Haemophilia A (without inhibitors)
Shaded boxes represent evidence-based information from either current systematic
review or from evidence cited in previous consensus-based guidelines (Australian
Haemophilia Centre Directors' Organisation 2004), unshaded boxes represent data from
product information sheets, current as at January 2005.
Table 51.
A matrix of treatment recommendations for patients with haemophilia A (without inhibitors
to FVIII)
Indication
Product
Mild
Acute
bleeding
DDAVP
Mild to moderate haemophilia:
Test dose for efficacy
0.3 µg/kg in 50 ml 0.9% saline infused
intravenously or subcutaneously over ≥
30 minutes
Use with caution in the
elderly
Moderate
Severe
Precautions
Contraindicated in patients
with arteriovascular disease
and in children < 2 years
One dose in 24 hours (≤3 consecutive
days).
Restrict fluid intake
rFVIII
pdFVIII
Minor
haemorrhage:
Moderate
haemorrhage:
Severe
haemorrhage:
Require peak
post-infusion FVIII
activity in the
blood:
Require peak
post-infusion FVIII
activity in the
blood:
Require peak
post-infusion FVIII
activity in the
blood:
20–40 IU/dL
30–60 IU/dL
60–100 IU/dL
Infuse every 12 to
24 hours for 1–3
days until bleeding
has stopped, pain
resolved or
healing achieved
Infuse every 12 to
24 hours for 3
days or more until
pain and disability
resolved
Repeat infusions
every 8–24 hours
until resolved
Minor
haemorrhage:
Moderate-severe haemorrhage:
Require peak
post-infusion FVIII
activity in the
blood:
20–30 IU/dL
Dose: 10–15 IU/kg
Infused 1–2 times
a day, for 1–2
days
164
Require peak post-infusion FVIII activity
in the blood:
30–80 IU/dL
Dose: 15–40 IU/kg
Infused 1–3 times a day, for 1–2 days or
7–10 days for intracranial haemorrhage
Appropriate laboratory tests
should be performed to
ensure appropriate levels of
FVIII in patient plasma are
achieved and maintained
If FVIII levels fail to reach
expected levels or bleeding
continues, the presence of
inhibitors should be
suspected
Inhibitor testing may be
performed within 20
exposure days after initial
dose
When factor concentrates
are used, recombinant
factors are preferred rather
than plasma-derived, due to
their higher safety profile
regarding transmission of
blood-borne agents
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 51
(cont.)
Tranexamic
acid
Standard dose:
Contraindications:
2–3 tablets of 0.5 g taken 2–3 times daily
Active thromboembolic disease
such as deep vein thrombosis,
pulmonary embolism and
cerebral thrombosis
Epistaxis
1.5 g orally 3 times daily for 4–10 days
Solution may be applied topically to the nasal mucosa
by soaking gauze strip in solution and packing nasal
cavity
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Patients with a high risk of
thrombosis (family history or
previous thrombotic event)
should be closely monitored
Should not be used with FEIBA
or other prothrombin complex
concentrates
Prophylaxis
rFVIII
Recommended dose range for FVIII prophylaxis is
25–40 IU/kg, 3 times per week or more frequently as
required
Infant delivery
Genetic
counselling
Should be accessible to all carriers of haemophilia,
including potential risks of antenatal testing and
potential complications during pregnancy and delivery
for both the foetus and mother
Antenatal care
Antenatal testing options depend on stage of
pregnancy
Chorionic villus sampling is the preferred method
Factor replacement may be required for invasive
procedures
Delivery of
infants
Vaginal delivery should be considered for all infants
with or suspected of haemophilia unless obstetric
factors indicate caesarean section
Prophylaxis in patients with
severe haemophilia should only
be started after bleeding
episodes have commenced,
due to possible increased risk
of inhibitor development with
administration in very young
children
Mother
Although haemostatic support
is rarely necessary, there is a
slight risk of bleeding in first
trimester and 3–5 days after
delivery. Therefore, FVIII:C
levels should be monitored
Instrument delivery should be
avoided (forceps and vacuum
extraction), as should fetal
scalp electrodes
Early recourse to caesarean is
recommended when there is a
failure of labour to progress
Cord blood samples or peripheral blood samples may
be taken from male infants of haemophilia carriers
Infant
Heel sticks and venipuncture
should be avoided
Vitamin K injections should be
avoided in affected male
infants; oral vitamin K may be
used as an alternative
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
165
Table 51
(cont.)
rFVIII
Surgery
Minor surgery:
Major surgery:
Require peak post-infusion
FVIII activity in the blood:
Require post-infusion FVIII
activity in the blood (preand post-operative):
60–80 IU/dL
pdFVIII
Tranexamic
acid
Single infusion plus oral
antifibrinolytic therapy for 1
hour, repeat if required
80–100 IU/dL
Minor surgery:
Major surgery
Loading:
Loading:
Require peak post-infusion
FVIII activity in the blood:
Require peak post-infusion
FVIII activity in the blood:
50–60 IU/dL
80–100 IU/dL
Dose: 20–30 IU/kg
Dose: 40–50 IU/kg
Maintenance:
Maintenance:
Require peak post-infusion
FVIII activity in the blood:
Require peak post-infusion
FVIII activity in the blood:
20–50 IU/dL
20–100 IU/dL
Dose: 15–30 IU/kg
Dose: 10–40 IU/kg
Repeat infusions every 8–
24 hours according to the
rate of healing
Intravenous dose: 10 mg/kg 2–3 times per day
(intravenous preparation available in Australia through
Special Access Scheme)
Oral dose: 25 mg/kg 2–3 times per day
Total doses:
80–100 mg/kg/day given orally
Paediatric dose 35 mg/kg/8hours
Appropriate laboratory tests
should be performed to
ensure appropriate levels of
FVIII in patient plasma is
achieved and maintained
If FVIII levels fail to reach
expected levels or bleeding
continues, the presence of
inhibitors should be suspected
When factor concentrates are
used, recombinant factors are
preferred rather than plasmaderived, due to their higher
safety profile regarding
transmission of blood-borne
agents
Contraindications:
Active thromboembolic
disease such as deep vein
thrombosis, pulmonary
embolism and cerebral
thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Patients with a high risk of
thrombosis (family history or
previous thrombotic event)
should be closely monitored
Should not be used with
FEIBA or other prothrombin
complex concentrates
(cont.)
166
Use of recombinant and plasma-derived Factor VIII and IX
Table 51
(cont.)
Fibrin glue
Fibrin sealant should only be used topically
Dose depends on size to be treated
4 cm2= 0.5 mL
8 cm2= 1.0 mL
16 cm2= 2.0 mL
40 cm2= 5.0 mL
Fibrin glue is derived from
human plasma, which may
contain infectious agents such
as viruses
Risk has been minimised
through donor screening and
inactivating known viruses, but
there is still a potential risk of
unknown infectious agents
Contraindicated in patients
hypersensitive to bovine proteins
Dental
Tranexamic
acid
10 mg/kg given intravenously (available through SAS in
Australia) prior to dental surgery
25 mg/kg given orally 3–4 times per day for 6–8 days
Contraindications:
Active thromboembolic disease
such as deep vein thrombosis,
pulmonary embolism and
cerebral thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Patients with a high risk of
thrombosis (family history)
should be closely monitored
Should not be used with FEIBA
or other prothrombin complex
concentrates
DDAVP = desmopressin; FEIBA = Factor eight inhibitor bypassing agent; SAS= Special Access Scheme
Use of recombinant and plasma-derived Factor VIII and IX
167
Haemophilia A (with inhibitors)
Shaded boxes represent evidence-based information from either current systematic
review or from evidence cited in previous consensus-based guidelines (Australian
Haemophilia Centre Directors' Organisation 2004), unshaded boxes represent data from
product information sheets, current as at January 2005.
Table 52.
Acute
bleeding
A matrix of treatment recommendations for patients with haemophilia A (with inhibitors to
FVIII)
Product
Mild-Moderate
Severe
rFVIIa
Early intervention doses of
90 µg/kg have been found
effective in treating mild–
moderate joint, muscle and
mucocutaneous bleeds
Major bleed in low-titre, high
responder
1–3 doses repeated at 3 hourly
intervals to achieve
haemostasis, and 1 additional
dose to maintain haemostasis
Dose interval may be
increased to 3–4 hours
Higher doses
required for
young
children
Mild bleed in low-titre, highresponder or high titre
90 µg/kg every 2 hours for a
minimum 2 doses, followed by
further dose
If response is not observed,
treatment with aPCCs should be
considered
Precautions
90 µg/kg every 2 hours for 12
hours or until clinical response
is observed
If response is not observed,
treatment with aPCCs should
be considered
High titre
90 µg/kg every 2 hours for 12
hours, increase to every 3
hours depending on clinical
response
Dose interval may be
extended to every 4 hours
Therapy may be continued to
14 days
Alternative schedule is
320 µg/kg every 6 hours
rFVIII
Mild bleed in low-titre, lowresponder
Major bleed in a low-titre, lowresponder
50–100 IU/kg repeated every 8–
12 hours
50–150 IU/kg repeated every
8–12 hours
Levels maintained and
monitored at >50% until
healing completed
aPCCs
Joint haemorrhage
FEIBA
50–100 IU/kg 2 times per day until clear signs of clinical
improvement, reduction of swelling or mobilisation of joint
Mucous membrane bleeding
50–100 IU/kg at 6 hourly intervals, not exceeding 200 IU/kg/day
Soft tissue haemorrhage
Monitor for side effects
which, while rare, are
serious
Patients should be
monitored for
thrombotic
complications
100 IU/kg at 12 hourly intervals, not exceeding 200 IU/kg/day
Other severe haemorrhages
100 IU/kg at 12 hourly intervals
168
Mild bleed in low-titre, highresponder or high titre
Major bleed in a low-titre,
high-responder or high titre
60–80 units/kg FEIBA as a
single dose
60–100 units/kg FEIBA two
times per day (maximum 200
units/kg/day)
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
Table 52
(cont.)
Plasmapheresis
Monitor for possible
anaphylactic reactions
Immunosuppression
Cyclophosphamide immunosuppressants not recommended
(future guidelines may include other new immunosuppressants
(e.g. Rituximab), but currently not enough evidence is
available)
Tranexamic
acid
Standard dose:
Contraindications:
2–3 tablets of 0.5 g taken 2–3 times daily
Active thromboembolic
disease such as deep
vein thrombosis,
pulmonary embolism
and cerebral thrombosis
Epistaxis
1.5 g orally 3 times daily for 4–10 days
Solution may be applied topically to the nasal mucosa by
soaking gauze strip in solution and packing nasal cavity
Subarachnoid
haemorrhage
Hypersensitivity to
tranexamic acid or any
of the ingredients
Precautions:
Patients with a high risk
of thrombosis (family
history or previous
thrombotic event)
should be closely
monitored
Should not be used with
FEIBA or other
prothrombin complex
concentrates
Surgery/
Dental
rFVIIa
An initial dose of 90 µg/kg immediately prior to surgery
Repeat dose after 2 hours, then 2–3 hour intervals for 24–48
hours
In major surgery dosage should continue every 2–4 hours for
6–7 days, dosage interval may then increase to every 6–8
hours for another 2 weeks
Major bleed in low-titre, high responder
90 µg/kg every 2 hours for 12 hours or until clinical response is
observed
Dose interval may be increased to 3–4 hours
If response is not observed, treatment with aPCCs should be
considered
High titre
90 µg/kg every 2 hours for 12 hours, increase to every 3 hours
depending on clinical response
Dose interval may be extended to every 4 hours
Therapy may be continued to 14 days
Alternative schedule is 320 µg/kg every 6 hours
Use of recombinant and plasma-derived Factor VIII and IX
(cont.)
169
Table 52 (cont.)
rFVIII
Surgery in a low-titre, low-responder
50–150 IU/kg repeated every 8–12 hours
Monitor and maintain levels at >50% until healing
completed
FEIBA
Surgery in a low-titre, high-responder or high titre
60–100 units/kg FEIBA two times per day (maximum 200
units/kg/day)
FVIII may be used for
patients with low- titre,
low-responding inhibitors
but should be avoided in
high-responding inhibitors
Monitor for side effects;
rare, but potentially
serious
Patients should be
monitored for thrombotic
complications
Tranexamic
acid
10 mg/kg given intravenously (available through SAS in
Australia) prior to dental surgery
25 mg/kg given orally 3–4 times per day for 6–8 days after
dental surgery
Intravenous dose: 10 mg/kg 2–3 times per day (available
through SAS in Australia)
Oral dose: 25 mg/kg 2–3 times per day
Paediatric dose 35 mg/kg/8hours
Contraindications:
Active thromboembolic
disease such as deep
vein thrombosis,
pulmonary embolism and
cerebral thrombosis
Subarachnoid
haemorrhage
Hypersensitivity to
tranexamic acid or any of
the ingredients
Precautions:
Patients with a high risk
of thrombosis (family
history) should be closely
monitored
Should not be used with
FEIBA or other
prothrombin complex
concentrates
Fibrin glue
Fibrin sealant should only be used topically
Dose depends on size to be treated
4 cm2= 0.5 mL
8 cm2= 1.0 mL
16 cm2= 2.0 mL
40 cm2= 5.0 mL
Fibrin glue is derived from
human plasma, which
may contain infectious
agents such as viruses
Risk has been minimised
through donor screening
and inactivating known
viruses, but there is still a
potential risk of unknown
infectious agents
Contraindicated in
patients hypersensitive to
bovine proteins
Tolerisation
rFVIII
Patients eligible for an international randomised controlled
trial comparing high and low dose protocols should
participate
Doses range from 25 IU/kg 2–3 times per week to 100
IU/kg/day
Tolerisation should continue until FVIII recovery > 60%,
and normal FVIII half life
aPCCs = activated prothrombin complex concentrates; FEIBA = Factor eight inhibitor bypassing agent; SAS= Special Access Scheme
170
Use of recombinant and plasma-derived Factor VIII and IX
Haemophilia B (without inhibitors)
Shaded boxes represent evidence-based information from either current systematic
review or from evidence cited in previous consensus-based guidelines (Australian
Haemophilia Centre Directors' Organisation 2004), unshaded boxes represent data from
product information sheets, current as at January 2005.
Table 53.
Acute
bleeding
A matrix of treatment recommendations for patients with haemophilia B (without inhibitors to
FIX)
Product
Mild
Moderate
Severe
Precautions
rFIX
Uncomplicated
haemarthroses,
superficial
muscular or soft
tissue:
Intramuscle or soft
tissue with
dissection,
mucous
membranes,
dental extractions,
haematuria:
Pharynx,
retropharynx,
retroperitoneum:
Patients transferred from
pdFIX may need higher
dose of rFIX:
Circulating FIX
activity required:
Ratio of pdFIX:rFIX:
≤15 years = 1:1.6
>15 years = 1:1.2
Circulating FIX
activity required:
Infuse every 12–
24 hours for 7–10
days
Circulating FIX
activity required:
20–30 IU/dL
Infuse every 12–
24 hours for 1–2
days
pdFIX
Desired plasma
concentration of
factor IX:
25–50 IU/dL
50-100 IU/dL
Infuse every 12–
24 hours for 2–7
days or until
bleeding stops
and healing
begins
Desired plasma concentration of factor
IX:
30–50 IU/dL
20–30 IU/dL
Dose: 30–50 IU/kg
Dose: 20–30 IU/kg
Infuse 1–2 times per day for 1-5 days
Infuse 1 time per
day for 1–2 days
Tranexamic
acid
An assay should be used to
monitor factor levels
When factor concentrates
are used, recombinant
factors are preferred rather
than plasma-derived, due to
their higher safety profile
regarding transmission of
blood-borne agents
Standard dose:
Contraindications:
2–3 tablets of 0.5 g taken 2–3 times daily
Active thromboembolic
disease such as deep vein
thrombosis, pulmonary
embolism and cerebral
thrombosis
Epistaxis
1.5 g orally 3 times daily for 4–10 days
Solution may be applied topically to the nasal mucosa by
soaking gauze strip in solution and packing nasal cavity
Subarachnoid haemorrhage
Hypersensitivity to
tranexamic acid or any of
the ingredients
Precautions:
Patients with a high risk of
thrombosis (family history)
should be closely monitored
Should not be used with
FEIBA or other prothrombin
complex concentrates
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
171
Table 53
(cont.)
rFIX
Prophylaxis
Infant
delivery
64–96 IU/kg three times per week for patients ≤15 years,
more frequently if required
48–72 IU/kg three times per week for patients >15 years,
more frequently if required
Genetic
counselling
Should be accessible to all carriers of haemophilia, including
potential risks of antenatal testing and potential complications
during pregnancy and delivery for both the foetus and mother
Antenatal
care
Antenatal testing options depend on stage of pregnancy
Prophylaxis should only be
started after bleeding episodes
have commenced, due to
current debate that there is an
increased risk of inhibitor
development with
administration in very young
children
Chorionic villus sampling is the preferred method
Factor replacement may be required for invasive procedures
Women with low FIX may require rFIX to control
haemorrhage
FIX:C should be measured soon after pregnancy is confirmed
and monitored in third trimester and postpartum
Delivery of
infants
Vaginal delivery should be considered for all infants with or
suspected of haemophilia unless obstetric factors indicate
caesarean section
Mother
Instrument delivery should be
avoided (forceps and vacuum
extraction) as should fetal
scalp electrodes
Early recourse to caesarean is
recommended when there is a
failure of labour to progress
Cord blood samples or peripheral blood samples may be
taken from male infants of haemophilia carriers
Infant
Heel sticks and venipuncture
should be avoided
Vitamin K injections should be
avoided in affected male
infants; oral vitamin K may be
used as an alternative
Surgery/
Dental
rFIX
Circulating FIX activity required:
50–100 IU/dL
Infuse every 12–24 hours for 7–10 days
pdFIX
Tranexamic
acid
10 mg/kg given intravenously (available through SAS in
Australia) prior to dental surgery
25 mg/kg given orally 3–4 times per day for 6–8 days after
dental surgery
Intravenous dose: 10 mg/kg 2–3 times per day (available
through SAS in Australia)
Oral dose: 25 mg/kg 2–3 times per day
When factor concentrates are
used, recombinant factors are
preferred rather than plasmaderived, due to their higher
safety profile regarding
transmission of blood-borne
agents
Contraindications:
Active thromboembolic disease
such as deep vein thrombosis,
pulmonary embolism and
cerebral thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Paediatric dose: 35 mg/kg/8hours
Patients with a high risk of
thrombosis (family history)
should be closely monitored
Should not be used with FEIBA
or other prothrombin complex
concentrates
(cont.)
172
Use of recombinant and plasma-derived Factor VIII and IX
Table 53
(cont.)
Fibrin glue
Fibrin sealant should only be used topically
Dose depends on size to be treated
4 cm2= 0.5 mL
8 cm2= 1.0 mL
16 cm2= 2.0 mL
40 cm2= 5.0 mL
Fibrin glue is derived from
human plasma, which may
contain infectious agents such
as viruses
Risk has been minimised
through donor screening and
inactivating known viruses, but
there is still a potential risk of
unknown infectious agents
Contraindicated in patients
hypersensitive to bovine
proteins
FEIBA = Factor eight inhibitor bypassing agent; SAS= Special Access Scheme
Use of recombinant and plasma-derived Factor VIII and IX
173
Haemophilia B (with inhibitors)
Shaded boxes represent evidence-based information from either current systematic
review or from evidence cited in previous consensus-based guidelines (Australian
Haemophilia Centre Directors' Organisation 2004), unshaded boxes represent data from
product information sheets, current as at January 2005.
Table 54.
Acute
bleeding
A matrix of treatment recommendations for patients with haemophilia B (with inhibitors to
FIX)
Product
Mild-moderate
Severe
rFVIIa
Early intervention
doses of 90 µg/kg have
been found effective in
treating mild–moderate
joint, muscle and
mucocutaneous bleeds
Initial dose of 90 µg/kg,
dosing every 2 hours
until clinical
improvement observed
1–3 doses repeated at
3 hourly intervals
achieve haemostasis,
and 1 additional dose
maintains haemostasis
If further treatment is
needed, dosing interval
may be extended to
every 3 hours for 1–2
days, and increased to
every 4, 6, 8, or 12
hours as judged
appropriate
aPCCs
Tranexamic
acid
Precautions
Patients should be closely
monitored for adverse events,
which though rare, may be
serious
Standard dose:
Contraindications:
2–3 tablets of 0.5 g taken 2–3 times daily
Active thromboembolic disease
such as deep vein thrombosis,
pulmonary embolism and cerebral
thrombosis
Epistaxis
1.5 g orally 3 times daily for 4–10 days
Solution may be applied topically to the nasal
mucosa by soaking gauze strip in solution and
package nasal cavity
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Patients with a high risk of
thrombosis (family history) should
be closely monitored
Should not be used with FEIBA or
other prothrombin complex
concentrates
(cont.)
174
Use of recombinant and plasma-derived Factor VIII and IX
Table 54 (cont.)
Tolerisation
rFIX
There is no information available on tolerisation
protocols for haemophilia B patients
Surgery/Dental
rFVIIa
An initial dose of 90 µg/kg immediately prior to
surgery
Repeat dose after 2 hours, then at 2–3 hourly
intervals for 24–48 hours
In major surgery dosage should continue every 2–4
hours for 6–7 days, then increase dosage interval
to every 6–8 hours for another 2 weeks
Fibrin glue
Fibrin sealant should only be used topically
Dose depends on size to be treated
4 cm2= 0.5 mL
8 cm2= 1.0 mL
16 cm2= 2.0 mL
40 cm2= 5.0 mL
Fibrin glue is derived from human
plasma, which may contain
infectious agents such as viruses
Risk has been minimised through
donor screening and inactivating
known viruses, but there is still a
potential risk of unknown
infectious agents
Contraindicated in patients
hypersensitive to bovine proteins
Tranexamic
acid
10 mg/kg given intravenously (available through
SAS in Australia) prior to dental surgery
25 mg/kg given orally 3–4 times per day for 6–8
days after dental surgery
Intravenous dose: 10 mg/kg 2–3 times per day
(available through SAS in Australia)
Oral dose: 25 mg/kg 2–3 times per day
Contraindications:
Active thromboembolic disease
such as deep vein thrombosis,
pulmonary embolism and cerebral
thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic
acid or any of the ingredients
Precautions:
Paediatric dose: 35 mg/kg/8hours
Patients with a high risk of
thrombosis (family history) should
be closely monitored
Should not be used with FEIBA or
other prothrombin complex
concentrates
aPCCs = activated prothrombin complex concentrates; FEIBA = Factor eight inhibitor bypassing agent; SAS= Special Access Scheme
Use of recombinant and plasma-derived Factor VIII and IX
175
von Willebrand disease
Shaded boxes represent evidence-based information from either current systematic
review or from evidence cited in previous consensus-based guidelines (Australian
Haemophilia Centre Directors' Organisation 2004), unshaded boxes represent data from
product information sheets, current as at January 2005.
Table 55.
A matrix of treatment recommendations for patients with von Willebrand disease
Indication
Product
Recommendations
Precautions
Acute bleeding
DDAVP
0.3 µg/kg in 50ml 0.9% saline infused
intravenously or subcutaneously over ≥
30 minutes
Test dose for efficacy
One dose in 24 hours (≤3 consecutive
days).
Use with caution in the elderly
Contraindicated in patients with
arteriovascular disease and in children < 2
years
Restrict fluid intake
pdFVIII/vWF
concentrate
Patients unresponsive to (or
contraindicated for) DDAVP should
receive pdFVIII/vWF concentrate
Tranexamic
acid
2–3 tablets 0.5 g, 2–3 times a day,
taken with water
Contraindications:
Active thromboembolic disease such as deep
vein thrombosis, pulmonary embolism and
cerebral thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic acid or any of
the ingredients
Precautions:
Patients with a high risk of thrombosis (family
history or previous thrombotic event) should
be closely monitored
Menorrhagia
Tranexamic
acid
Start treatment when bleeding is first
noticed
Take 2–3 tablets 4 times a day for 4
days
Swallow with water
If menstrual bleeding is not reduced by
tranexamic acid, alternative therapy
should be considered
If menstrual bleeding is irregular, the cause
of irregularity should be established prior to
use of tranexamic acid
Contraindications:
Active thromboembolic disease such as deep
vein thrombosis, pulmonary embolism and
cerebral thrombosis
Subarachnoid haemorrhage
Hypersensitivity to tranexamic acid or any of
the ingredients
Precautions:
Patients with a high risk of thrombosis (family
history or previous thrombotic event) should
be closely monitored
(cont.)
176
Use of recombinant and plasma-derived Factor VIII and IX
Table 55 (cont.)
DDAVP
0.3 µg/kg in 50 ml 0.9% saline infused
intravenously or subcutaneously over ≥
30 minutes
One dose in 24 hours (≤3 consecutive
days).
Test dose for efficacy
Use with caution in the elderly
Contraindicated in patients with
arteriovascular disease and in children < 2
years
Restrict fluid intake
Oestrogens
Infant delivery
DDAVP
Women with Type 1 vWD may require
DDAVP in the first 3–4 days after
delivery
Instrument delivery should be avoided
(forceps and vacuum extraction) as should
fetal scalp electrodes
Genetic
counselling
Should be accessible to all women with
vWD, including potential risks of
antenatal testing and potential
complications during pregnancy and
delivery for both the foetus and mother
Early recourse to caesarean is
recommended when there is a failure of
labour to progress
Antenatal testing options depend on
stage of pregnancy
Vitamin K injections should be avoided in
affected infants; oral vitamin K may be
used as an alternative
Antenatal care
Chorionic villus sampling is the
preferred method
Heel sticks and venipuncture should be
avoided
Factor concentrates may be required for
invasive procedures
Surgery/Dental
DDAVP
0.3 µg/kg in 50 ml 0.9% saline infused
intravenously or subcutaneously over ≥
30 minutes
One dose in 24 hours (≤3 consecutive
days).
Test dose for efficacy
Use with caution in the elderly
Contraindicated in patients with
arteriovascular disease and in children < 2
years
Restrict fluid intake
pdFVIII/vWF
concentrate
Tranexamic
acid
To be used in conjunction with DDAVP
or FVIII/vWF
10 mg/kg given intravenously (available
through SAS in Australia) prior to dental
surgery
Contraindications:
Active thromboembolic disease such as
deep vein thrombosis, pulmonary
embolism and cerebral thrombosis
Subarachnoid haemorrhage
25 mg/kg given orally 3-4 times per day
for 6–8 days after dental surgery
Hypersensitivity to tranexamic acid or any
of the ingredients
Intravenous dose: 10 mg/kg 2–3 times
per day (available through SAS in
Australia)
Precautions:
Oral dose: 25 mg/kg 2–3 times per day
Patients with a high risk of thrombosis
(family history or previous thrombotic
event) should be closely monitored
Paediatric dose: 35 mg/kg/8hours
Fibrin glue
To be used in conjunction with DDAVP
or pdFVIII/vWF
Fibrin sealant should only be used
topically
Dose depends on size to be treated
4 cm2= 0.5 mL
8 cm2= 1.0 mL
16 cm2= 2.0 mL
Fibrin glue is derived from human plasma,
which may contain infectious agents such
as viruses
Risk has been minimised through donor
screening and inactivating known viruses,
but there is still a potential risk of unknown
infectious agents
Contraindicated in patients hypersensitive
to bovine proteins
40 cm2= 5.0 mL
DDAVP = desmopressin; FEIBA = Factor eight inhibitor bypassing agent; pdFVIII/vWF concentrate = plasma-derived factor concentrate
enriched with von Willebrand factor; SAS= Special Access Scheme
Use of recombinant and plasma-derived Factor VIII and IX
177
Appendix E
Methodology
Inclusion criteria
Studies were included for each area of the guidelines (e.g. management of the disease,
delivery of infants), if they addressed the population, therapy, comparator, outcomes,
study design, search period and language delineated in Box 1.
Box 1.
Study selection criteria
Selection criteria
Inclusion criteria
Population
Management of the diseases
All patients with clinically diagnosed haemophilia A, haemophilia B, von Willebrand disease, or other
rare bleeding disorders.
Treatment of bleeding episodes/Prophylaxis and treatment/Adjunctive haemostatic agents
All patients with clinically diagnosed haemophilia A, haemophilia B, von Willebrand disease, or other
rare bleeding disorders (including those undergoing surgery or dental care).
Delivery of infants
Pregnant women who are carriers of haemophilia A or haemophilia B or with von Willebrand Disease.
Infants with haemophilia A, B or von Willebrand Disease.
Tolerisation procedures
Patients with haemophilia A or B with inhibitors to factor VIII or IX.
Therapy
Management of the diseases
Haemophilia A or B: Management of patients using recombinant factor VIII or IX.
Von Willebrand disease: Management of patients using plasma-derived factor VIII.
Treatment of bleeding episodes
Haemophilia A or B: On-demand treatment of patients with recombinant factor VIII or IX.
Von Willebrand disease: On-demand treatment of patients using plasma-derived factor VIII with von
Willebrand factor.
Prophylaxis and treatment of the diseases
Haemophilia A or B: Prophylaxis and treatment of patients using recombinant factor VIII or IX (including
before, during and after surgery, dental care and other situations where there is a risk of bleeding).
Von Willebrand disease: Prophylaxis and treatment of patients using plasma-derived factor VIII
(including before, during and after surgery, dental care and other situations where there is a risk of
bleeding).
Adjunctive haemostatic agents
Treatment of patients with adjunctive haemostatic agents, such as antifibrinolytic agents (e.g.
aminocaproic acid, tranexamic acid), desmopressin, and topical applications (e.g. thrombin and fibrin
sealant).
Delivery of infants
Haemophilia A or B: Prophylactic or on-demand treatment with recombinant factor VIII or IX ante-, periand postnatally.
Von Willebrand disease: Prophylactic or on-demand treatment with plasma-derived factor VIII ante-,
peri- and postnatally.
Tolerisation procedures
Treatment with regular administration of recombinant factor VIII or IX.
178
(cont.)
Use of recombinant and plasma-derived Factor VIII and IX
(cont.)
Comparator
Haemophilia A or B: ‘Usual’ or standard care of patients using appropriate plasma-derived factorsa.
Von Willebrand disease: Standard care of patients using desmopressin (mild) or concentrated von
Willebrand factor (severe)
Outcomesb
Primary: mortality (survival), morbidity (including transmission of viral infections, thrombosis, myocardial
infarction), number and severity of bleeding episodes (including clotting activity, recovery/response to
factor), level of factor, development/level of inhibitors
Secondary: Quality of life, hospitalisation, length of treatment (including number of infusions required,
number of exposure days), dose required for haemostasis control
Study design
Effectiveness: Systematic reviews, randomised controlled trials, non-randomised controlled trials, cohort
studies, before-and-after controlled studies, case-control studies. When high quality evidence was not
available, large case series of consecutive patients, analysed on an intention-to-treat basis, were
included.
Safety: Systematic reviews, randomised controlled trials, non-randomised controlled trials, cohort
studies, before-and-after controlled studies, case-control studies, case-series
Search Period
1966 to September 2004
Language
Studies in languages other than English were translated and included if they represented a higher level
of evidence than that available in the English literature evidence-base.
Limitations
Human
All children <18 years old and adults who have not been infected with a virus have been using recombinant factors only. From 1 October
2004, all haemophilia patients were given access to recombinant products (Department of Health and Ageing 2004); b Objective outcome
measures are preferred to subjective outcome measures
a
Search strategies
The electronic sources listed in Table 56 were searched for potentially relevant literature.
Other sources of literature (Table 57) were also searched.
Table 56.
Bibliographic databases
Electronic database
Time period
AustHealth
1997 to September 2004
Cinahl
1977 to September 2004
Cochrane Library—including Cochrane Database of
Systematic Reviews, Database of Abstracts of Reviews of
Effects, the Cochrane Central Register of Controlled Trials
(CENTRAL), the Health Technology Assessment Database,
the NHS Economic Evaluation Database
1966 to September 2004
Current Contents
1993 to September 2004
Embase.com (includes Medline and Embase)
1966 to September 2004
Pre-Medline
1966 to September 2004
ProceedingsFirst
1993 to September 2004
PsycInfo
1983 to September 2004
Web of Science—Science Citation Index Expanded
1995 to September 2004
Use of recombinant and plasma-derived Factor VIII and IX
179
Table 57.
Other sources of evidence (1966 to September 2004)
Electronic database
Time period
Australian Department of Health and Ageing
http://www.health.gov.au/
Current Controlled Trials metaRegister
http://controlled-trials.com/
Health Technology Assessment International
http://www.htai.org
International Network for Agencies for Health Technology
Assessment
http://www.inahta.org/
NHMRC— National Health and Medical Research Council
(Australia)
http://www.health.gov.au/nhmrc/
National Library of Medicine Health Services / Technology
Assessment Text
http://text.nlm.nih.gov/
National Library of Medicine Locator Plus database
http://locatorplus.gov
Trip database
http://www.tripdatabase.com
UK National Research Register
http://www.update-software.com/national/
US Department of Health and Human Services (reports and
publications)
http://www.os.dhhs.gov/
Websites of Haemophilia/Haematology Associations
See Appendix F
Other medical and health websites
See Appendix F
Hand searching (Journals from 2003–2004)
Blood reviews
Library or electronic access
Journal of thrombosis and haemostasis
Library or electronic access
Thrombosis and haemostasis
Library or electronic access
Clinical and laboratory haematology
Library or electronic access
Haematology
Library or electronic access
Blood
Library or electronic access
Haemophilia
Library or electronic access
Annals of haematology
Library or electronic access
British journal of haematology
Library or electronic access
Transfusion
Library or electronic access
Vox sanguinis
Library or electronic access
Expert clinicians
Any information provided by expert clinicians associated with
this review was assessed as to whether it met the inclusion
criteria.
Clinical consultants
Pearling
All included articles had their reference lists searched for
additional relevant source material.
The search terms used—including text words and indexing (e.g. Emtree) headings—
when searching the electronic databases (Table 56) are given in Table 58.
180
Use of recombinant and plasma-derived Factor VIII and IX
Table 58.
Search terms utilised
Area of inquiry
Search terms
All searches
EMTREE
Hemophilia A; Hemophilia B; von Willebrand disease; blood clotting factor 8; blood clotting
factor 9
Text words
H*emophilia*; factor VIII; factor IX; factor 8; factor 9; von Willebrand disease; von Willebrands
disease; von Willebrand syndrome; von Willebrands syndrome
Management
EMTREE
Disease management; patient care; therapy
Text words
Disease management
Adjunctive haemostatic
agents
EMTREE
Aminocaproic acid; tranexamic acid; antifibrinolytic agent
Text words
Aminocaproic acid; tranexamic acid; thrombin; fibrin; desmopressin; antifibrinolytic agent
Inhibitor development
EMTREE
Blood clotting inhibitor; activated prothrombin complex
Text words
Inhibitor*; tabiliz*; toleri?ation; toleri?ed; tolerance induction; immune tolerance; FEIBA
Treatment of bleeding
episodes
EMTREE
Dental care; surgery; prophylaxis; bleeding
Text words
Dental; surgery; surgical; prophyla*; bleeding
Infant delivery
EMTREE
Labor; obstetric care; delivery
Text words
Labo*r; obstetric; delivery
Infectious diseases and
other safety issues
EMTREE
Hepatitis; acquired immune deficiency syndrome; life expectancy; complication; infection; side
effect; risk assessment; safety; bleeding; human immunodeficiency virus infection; adverse
drug reaction; mortality; morbidity
Text words
Safety; mortality; morbidity; AIDS; HIV; human immunodeficiency virus; haemorrhage; hepatitis;
bleeding; complication*; adverse event*
The process for selecting studies for this review and clinical guidelines document is
shown in Figure 2.
Use of recombinant and plasma-derived Factor VIII and IX
181
Figure 2.
Study selection process
1. All reference citations from all literature sources were
collated into an Endnote 6.0 database.
2. Duplicate references were removed.
3. Studies were excluded on the basis of the complete citation
information if it was obvious that they did not meet the
inclusion criteria. All other studies were retrieved for fulltext assessment.
4. Inclusion criteria were applied independently to the full-text
articles by one researcher. Those meeting the criteria formed
part of the evidence-base. The remainder provided
background information.
5. The reference lists of the included articles were pearled for
additional relevant studies. These were retrieved and
assessed according to phase 4.
6. The evidence-base consisted of articles from phases 4 and 5
that met the inclusion criteria.
Table 59 provides a breakdown of the study selection process in terms of the number of
literature citations or articles retrieved and retained from each phase of the process. Any
doubt concerning inclusions at Phase 4 was resolved by group consensus.
Table 59.
Number of citations initially retrieved and then retained at each phase
Phase 1
Phase 2
Phase 3
Phase 4
Phase 5
Phase 6
29 945
12 087
1220
439
14
96
(Total Included)
Database searches generated 29,945 citations published between 1966 and September
2004. After duplicates were removed, 12,087 citations were screened initially by two
reviewers who excluded articles that were clearly not relevant to this report. Articles
excluded immediately by title and abstract included narrative reviews, letters, comments
and opinions, editorials and individual case reports.
182
Use of recombinant and plasma-derived Factor VIII and IX
Four hundred and thirty-nine full-text articles were retrieved and provided data for this
review. Of these, 82 were included to assess effectiveness and safety issues. A further 14
studies were included after the reference lists of the included articles were pearled. The
total evidence base comprised 96 original studies and systematic reviews.
Use of recombinant and plasma-derived Factor VIII and IX
183
Appendix F
Specialty Websites
Haemophilia websites
•
Abstracts from Haemophilia—the Journal of WFH
http://www.blackwell-synergy.com
•
American Society of Hematology
http://www.hematology.org/
•
Association of Hemophilia Clinic Directors of Canada
www.ahcdc.ca/
•
Australasian Society of Thrombosis and Haemostasis
http://www.asth.org.au/
•
Australia and New Zealand Society of Blood Transfusion
http://www.anzsbt.org.au/
•
Australian Bleeding Disorders Registry
http://www.ahcdo.org.au/abdr
•
Australian Haemophilia Centre Directors’ Organisation
http://www.ahcdo.org.au/
•
British Society of Haematology
http://www.b-s-h.org.uk/
•
Haematology Society of Australia and New Zealand
http://www.hsanz.org.au/
•
Haemophilia Directors for Scotland and Northern Ireland
www.rcpe.ac.uk/esd/clinical_standards/hdsni/hdsni_index.html
•
Haemophilia Foundation ACT (HFACT)
http://www.hfact.org.au
•
Haemophilia Foundation Australia
http://www.haemophilia.org.au
•
Haemophilia Foundation Queensland (HFQ)
http://www.hfq.org.au
•
Haemophilia Foundation Victoria (HFV)
http://www.haemophiliavic.org.au
•
Haemophilia Society (UK)
http://www.haemophilia.org.uk
184
Use of recombinant and plasma-derived Factor VIII and IX
•
Hemophilia Galaxy
http://www.hemophiliagalaxy.com
•
Hemophilia Village
http://www.hemophilia-village.net
•
National Hemophilia Foundation (US)
http://www.hemophilia.org
•
New Zealand Haemophilia Foundation
http://www.haemophilia.org.nz
•
Oxford Haemophilia Centre
http://www.medicine.ox.ac.uk
•
Shemophilia (US)
http://www.shemophilia.org
•
South African Haemophilia Foundation
http://www.haemophilia.org.za
•
The British Committee for Standards in Haematology
http://www.bcshguidelines.com/
•
The Canadian Hemophilia Society
http://www.hemophilia.ca
•
The Haemophilia Alliance (UK)
http://www.haemophiliaalliance.org.uk
•
The International Society of Haemostasis and Thrombosis
http://www.med.unc.edu/isth/welcome
•
United Kingdom Haemophilia Centre Directors Organisation
www.medicine.ox.ac.uk/ohc/ukhcdo.htm
•
World Federation of Hemophilia
http://www.wfh.org
Use of recombinant and plasma-derived Factor VIII and IX
185
Other relevant medical and health websites
•
Australian Red Cross Blood Service
http://www.donateblood.com.au
•
Medicines Australia
http://www.medicinesaustralia.com.au/
•
National Blood Authority
http://www.nba.gov.au
•
Novo Seven
http://www.novoseven.com
•
World Health Organization
http://www.who.int/en/
186
Use of recombinant and plasma-derived Factor VIII and IX
Appendix G Consensus Clinical Practice
Guidelines
Previous Australian clinical guidelines on treatment and
prophylaxis
6.1. General recommendations (Haemophilia Foundation Australia Medical
Advisory Panel 2000)
6.1.1. Patient information and consent. Good practice dictates that the necessity for
treatment is appropriately explained to the patient and/or parent. This should include the
advantages and risks of different therapies to allow an informed decision to be made.
When consent has been obtained this should be recorded in the case notes.
6.1.2. Vaccination against hepatitis A and B. All patients who are not immune to
hepatitis A or B and who currently receive, or may require, blood products should be
vaccinated. At present revaccination with hepatitis A vaccine is not recommended and
the vaccine is not currently licensed for use in children under the age of one year.
Immunity to hepatitis B requires periodic reassessment and revaccination when
appropriate.
6.1.3. Risk reduction. The use of fractionated, virucidally treated, concentrates when
available has been the treatment of choice in achieving haemostasis in congenital
coagulation factor deficiency since these products carry a lower risk of transmitting
serious viral infection than cryoprecipitate or FFPcc (Grade B recommendation based on
Level III evidence). It is acknowledged that despite new viral inactivation techniques it is
possible that coagulation factor concentrates still transmit virus infection. For this reason
recombinant factor VIII is now preferred to treatment with plasma-derived concentrates.
6.1.4. Use of DDAVP (desamino-8-D-arginine vasopressin, desmopressin).
DDAVP should be considered for all patients with mild/moderate haemophilia A or
mild vWD, as this avoids the risk of viral transmission and is less expensive (Grade B
recommendation based on Level IIa evidence). DDAVP is generally administered
intravenously at a dose of 0.3 mcg/kg diluted in 50 mL of 0.9% saline and infused over
at least 30 minutes. An unlicensed intranasal spray preparation of DDAVP (Octim Nasal
Spray, Ferring) is availabledd at a dose of 300 mcg for adults and 150 mcg for children.
An unlicensed* concentrated subcutaneous preparation (Octim injection, Ferring) is also
available and should be given at the usual dose of 0.3 mcg/kg. Efficacy should be
demonstrated irrespective of the route employed, by measuring FVIII/vWF. Intranasal
DDAVP has been shown to be comparable to the effect of an intravenous dose of 0.2
cc
FFP—fresh frozen plasma
dd
Nasal spray is not routinely available for use (personal communication, Dr John Rowell)
*not registered under the Therapeutic Goods Act
Use of recombinant and plasma-derived Factor VIII and IX
187
mcg/kg DDAVP. DDAVP should be used with caution in elderly individuals, pregnant
women and avoided in those with evidence of arteriovascular disease. Precautions to
prevent fluid overload leading to hyponatraemia must be taken particularly in young
children and DDAVP is probably best avoided in those younger than two years of age.
6.1.5. Tranexamic acid. Tranexamic acid is an antifibrinolytic agent which
competitively inhibits the activation of plasminogen to plasmin and is available in an
intravenous or oral preparation (both in suspension and tablet form). The intravenous
preparation is not currently licensed* in Australia. Tranexamic acid is particularly useful
for bleeding from the gastrointestinal tract, menorrhagia, open wounds, dental surgery
and in conjunction with DDAVP (Grade A recommendation based on Level Ia
evidence). The recommended intravenous dose is 10 mg/kg 2–3 times daily and the oral
dose 25 mg/kg 2–3 times daily (Grade B recommendation based on Level IIa evidence).
Tranexamic acid is contra-indicated in patients with thromboembolic disease and should
be avoided in patients with haematuria. It should not be used with FEIBAee or other
prothrombin complex concentrates (Grade C recommendation based on Level IV
evidence). Tranexamic acid can be used in combination with NovoSevenff.
6.1.6 Aminocaproic acidgg. Aminocaproic acid is an antifibrinolytic agent which
competitively inhibits the activation of plasminogen to plasmin and is available as an
intravenous preparation. It is particularly useful for bleeding from the gastrointestinal
tract, menorrhagia, open wounds, dental surgery and in conjunction with DDAVP
(Grade A recommendation based on Level Ia evidence). The recommended intravenous
dose is 1 g hourly by continuous infusion. Aminocaproic acid is contraindicated in
patients with thromboembolic disease and should be avoided in patients with haematuria.
It should not be used with FEIBA or other prothrombin complex concentrates (Grade C
recommendation based on Level IV evidence). Aminocaproic acid can be used in
combination with rFVIIa.
6.2. Specific recommendations
Licensed* coagulation factor concentrates should be used in preference to unlicensed*
products. Each patient should be considered individually taking into account the
following recommendations.
6.2.1. Haemophilia A—factor VIII deficiency. For patients for whom coagulation
factor concentrate is the treatment of choice the following therapeutic strategies are
recommended. Recombinant factor VIII is the treatment of choice for all patients. As
ee
Factor Eight Inhibitor Bypassing Agent
ff
Recombinant human coagulation factor VIIa (rFVIIa)
gg Aminocaproic acid may become unavailable as the company, AMICAR, has ceased production (personal
communication, Dr Ben Saxon, Red Cross Blood Service)
* Registered under the Therapeutic Goods Act
† These recommendations have changed according to the 2004 government announcement to fund access
to recombinant clotting factors for all haemophilia patients. Access to recombinant factors is no longer
prioritised according to patient group.
188
Use of recombinant and plasma-derived Factor VIII and IX
the introduction of recombinant factor VIII has to be prioritised then those who may
benefit most should receive it first†.
6.2.2. von Willebrand disease. DDAVP should be used for DDAVP-responsive vWD
patients in preference to plasma-derived products. Where DDAVP is not likely to be
effective, or is contraindicated, FVIII concentrate or purified von Willebrand factor is
the treatment of choice. Cryoprecipitate is not virally inactivated and carries a risk of
virus transmission. It is, however, recognised that there are some circumstances in which
its use may be justified (Grade B recommendation based on Level Iib evidence).
6.2.3. Haemophilia B—factor IX deficiency. Patients with factor IX deficiency
should be treated with high-purity FIX concentrates because they cause less haemostatic
activation than PCCshh (Grade A recommendation based on Level Ib evidence) †.
6.2.4. Factor XI deficiency. The majority of patients with FXI:C levels < 15 u/dL will
suffer excessive bleeding following trauma or surgery and should be managed with
infusions of factor XI concentrate. In those with partial deficiency of factor XI (15–70
u/dL) bleeding is more difficult to predict. Where there is a clear history of abnormal
bleeding and haemostatic support is required, the use of FXI concentrate is justified. The
dose of FXI should be sufficient to raise the level of factor XI: C to 70 u/dL and the
level should not exceed 100 u/dL because of the risk of thrombosis (maximum dose 30
u/kg) (Grade C recommendation based on Level IV evidence). Where there is no history
of bleeding, tranexamic acid may be used alone, but in the event of subsequent excessive
bleeding must be replaced by FXI concentrate. Patients should be assessed for
preexisting risk of thrombosis and the concentrate should be used with great caution in
those with a history of cardiovascular disease (Grade C recommendation based on Level
IV evidence). FFP might be a suitable alternative when FXI concentrate is
contraindicated.
6.2.5. Factor VII deficiency. A purified, heat-treated FVII concentrate is available. This
should replace PCCs and FFP as the treatment of choice in situations in which
haemostatic support is necessary. The dose of FVII required depends on the severity of
the deficiency and on clinical circumstances. The level of FVII required for haemostasis
may be as low as 10–20 u/dL and this can be achieved by administering 5–10 i.u.
FVII/kg (Grade C recommendation based on Level IV evidence). Recombinant factor
VIIa can also be used in factor VII deficiency.
6.2.6. Factor II or X deficiency. There is currently no specific factor II or X
concentrate available and PCCs remain the treatment of choice (Grade C
recommendation based on Level IV evidence).
6.2.7. Factor V deficiency. There are no specific concentrates available for use in FV
deficiency and therefore FFP is the only available treatment (Grade C recommendation
based on Level IV evidence).
hh
Prothrombin complex concentrates
† These recommendations have changed according to the 2004 government announcement to fund access
to recombinant clotting factors for all haemophilia patients. Access to recombinant factors is no longer
prioritised according to patient group.
Use of recombinant and plasma-derived Factor VIII and IX
189
6.2.8. Factor XIII deficiency. FXIII concentrate prepared from human plasma is now
available and is the treatment of choice (Grade C recommendation based on Level IV
evidence).
6.2.9. Fibrinogen deficiency. Unlicensed* concentrates of fibrinogen have recently
become available and since these products are virally inactivated it is anticipated that they
may replace cryoprecipitate in the near future (Grade C recommendation based on Level
IV evidence).
6.2.10. Future treatment of hereditary coagulation disorders. Recombinant factor
VIII and IX will be used widely because of continuing concerns about the safety of
plasma-derived concentrates. It is anticipated that recombinant factor VIII, which is
formulated without addition of human albumin as a stabiliser, will become licensed.
Furthermore it is known that recombinant factor VIII being manufactured without using
any bovine or human proteins will eventually replace current products. Recombinant
factor IXii is currently licensed in the USA. It should be introduced for routine
haemophilia B care using criteria similar to those for recombinant factor VIII.
Clinical guidelines on prophylactic treatment using factors
VIII and IX
(Revised by the Clinical Advisory Sub Committee—Haemophilia, September 2000)
(AHMAC 2003)
1.
The goal of prophylaxis is to improve quality of life for patients with severe–
moderate haemophilia A and B by maintaining sufficient factor VIII and IX levels to
prevent spontaneous joint bleeding and the morbidity associated with complications
of joint bleeds.
2.
Factor VIII and factor IX prophylaxis is recommended for all children with severe
haemophilia A and B up the age of 18 years. If the supplies of factors VIII and IX
permit, consideration should be given to extending prophylaxis treatment to severe
haemophilia A and B patients older than 18 years.
3.
For the purposes of these guidelines, children have severe haemophilia when they
have major spontaneous bleeds into joints and their factor VIII and IX levels are
less than 5%.
4.
The recommended dosage range for factor VIII prophylaxis is 25–40 IU per kilo,
three times a week or more frequently as required. The recommended dosage for
factor IX prophylaxis is 40–60 IU per kilo, twice a week or more frequently as
required. The amount of factors VIII and IX used for prophylaxis will vary from
Recombinant factor IX has been registered for use in Australia since 2000 and funding to supply
commenced in 2001 (personal communication, Sharon Caris, Haemophilia Foundation Australia)
ii
* not registered under the Therapeutic Goods Act
190
Use of recombinant and plasma-derived Factor VIII and IX
patient to patient but for each patient will be the minimum amount of factor VIII or
IX required to prevent spontaneous joint bleeding. Please note that the guidelines
for factor IX dosage are based on plasma concentratesjj.
5.
The age at which prophylaxis therapy is introduced will vary from one to two years
to around five years depending on the number and severity of bleeds and whether
the patient and his family are willing to comply with the prophylactic treatment
regime.
6.
The recommended priority groups* for receiving recombinant factor VIII and factor
IX for treatment/prophylaxis, are in the following order of priority:
a. Previously untreated patients (PUPs)
b. Patients who have been previously treated with plasma-derived factor VIII and
factor IX but who have no evidence of hepatitis B, hepatitis C or HIV infection
7.
The recommended priority groups* for receiving recombinant factors VIII and IX
for treatment/prophylaxis (as supplies become available) in addition to the above
groups are, in order of priority:
a. virally infected children (up to 18 years); and
b. virally infected adults
Guidelines for the treatment of inhibitors in haemophilia A
Developed by AHCDO (Australian Haemophilia Centre Directors' Organisation 2004).
General comments
The diagnosis of an inhibitor, irrespective of titre, requires active monitoring and
treatment to determine whether the inhibitor progresses to a high titre inhibitor or is
transient. The diagnosis of an inhibitor should be conducted using Bethesda criteria.
After initial diagnosis of a low titre inhibitor in a patient with haemophilia A, routine
doses of factor VIII may be as much as doubled and the clinical response and inhibitor
titre monitored. Subsequent poor clinical response or rising titre levels indicate a nontransient inhibitor. The diagnosis of a high titre inhibitor, which does not respond to
factor VIII, should be treated with an alternative product.
The dosage for prophylaxis with recombinant factors are: factor VIII—1:1 (plasma-derived:recombinant)
factor IX—1:1.25 (plasma-derived:recombinant)
jj
* Access to recombinant factors is no longer prioritised according to patient group.
Use of recombinant and plasma-derived Factor VIII and IX
191
There is an incidence of inhibitors in those with mild haemophilia A following treatment
with factor VIII concentrates and the guidelines will also apply to those patients.
DDAVP is occasionally used in mild to moderate haemophilia but is often ineffective.
Minor bleeds include muscle and joint haemarthrosis but are not trauma related. Major
bleeds include trauma related muscle or joint injury, severe spontaneous muscle bleeds
e.g. psoas or iliacus, and haemorrhages affecting an organ.
Surgery, which affects inhibitor patients, includes dental procedures, insertion of
intravenous access devices and emergency and major elective procedures. Both dental
surgery and insertion of intravenous access devices require shorter durations of
replacement therapy.
Products available
3.1.1. Factor VIII: In the case of life threatening haemorrhage, infusion of factor VIII in
large doses can be used to swamp the inhibitor. This therapy is mostly used in patients
with low responding inhibitors (inhibitor titre is <5 BU/mL after infusion of factor
VIII). In patients who are high responders, this treatment may be effective provided the
inhibitor level is less than 5 BU/mL. Factor VIII levels should be observed to assess and
monitor response. Anamnesis can occur 5–7 days after therapy and make factor VIII
ineffective. Recombinant and plasma-derived factor VIII are available.
3.1.2. Recombinant Factor VII: This product has been widely used in Australia and has
proved to be highly effective in the management of spontaneous bleeding episodes,
which are life or limb threatening. Evidence in the literature suggests that it is effective in
79–92% of such episodes. In addition, there is evidence that it is effective in over 90% of
cases of surgery. Recombinant factor VIIa is infused as a bolus. Continuous infusion of
recombinant factor VIIa may reduce the quantity and cost of treatment but evidence is
conflicting. A recent study suggests continuous infusion of 50 µg/kg/hr is effective in
surgery. Antifibrinolytics are administered concurrently. The standard adult dose of
recombinant factor VIIa is 90 µg/kg. However, in children the mean half-life is
substantially reduced to 1.32 hours and thus higher doses of up to 200–250 µg/kg may
be required. Present indications for funding of recombinant factor VIIa by the National
Blood Authority (NBA) are for limb and life threatening haemorrhages. There is
widespread evidence that recombinant factor VIIa is effective for treatment of all
haemorrhage in patients with high responding factor VIII inhibitors. The appropriate
treatment for all haemorrhages should be extended to all children and adults regardless
of their state of residency.
3.1.3. Activated Prothrombin Complex Concentrates (APCCs) eg FEIBA VH:
Activated prothrombin complex concentrate, such as FEIBA VH, is effective in the
treatment of 90% of bleeding episodes, and has been effective in the management of
bleeding during major surgery. An effective dose is 60–100 units/kg twice per day. The
maximum daily dose of FEIBA is 200 units/kg/day. Antifibrinolytic agents, such as
tranexamic acid, should not be administered concurrently with FEIBA. It should be
noted that FEIBA contains small amounts of factor VIII and therefore may cause
elevation of inhibitor titres in some patients. The 1997 Working Party Report did not
recommend the use of these agents as first choice because they are plasma-derived rather
than recombinant products, there is a reported high incidence of thrombosis associated
with their use, and it is not possible to measure their activity in a standardised way.
192
Use of recombinant and plasma-derived Factor VIII and IX
Nevertheless, AHCDO recommends that these products remain an option for treatment
for complex cases in which alternative methods have proved ineffective. In patients who
are having frequent bleeds, a trial of FEIBA as prophylaxis should be considered. The
suggested dose is 75–100 units/kg three times a week.
3.1.4. Prothrombinex-HT: Despite general scepticism about the effectiveness of
prothrombinex-HT in the management of joint haemorrhage, some patients report
benefit and continue to be treated with this product. There are concerns about the
incidence of thrombosis when using repeated high doses, particularly in the presence of
liver disease and when used in combination with antifibrinolytics. There is no evidence of
efficacy in serious haemorrhages in surgery. AHCDO recommends that recombinant
factor VIIa is used in future for these patients.
3.1.5. Antifibrinolytic therapy: The recommended dose of tranexamic acid is 80–100
mg/kg/day, with a standard dose being 1 g qid given orally (recommended paediatric
dose is 35 mg/kg/8hr). An intravenous preparation is available in Australia through the
SAS. Intravenous epsilon aminocaproic acid (EACA) is no longer produced and not
available.
3.1.6. Plasmapheresis/Immunoadsorption: Plasmapheresis can be used to reduce
inhibitor titres to allow effective therapy with factor VIII. Specific immunoadsorption
using the Malmö protocol is not available in Australia. There is as yet little information
on the use of immunosuppression in patients with factor VIII inhibitors but some
experimental protocols are being proposed. Immunosuppression has been associated
with side effects including delayed wound healing and increased susceptibility to
infection. Rituximab (anti CD 20 monoclonal antibody) therapy may be considered as an
adjunct therapy to reduce inhibitor titres.
Treatment regimens
3.2.1. Low titre inhibitor (<5 BU/ml), low responder
Minor bleeding: The recommended dose of factor VIII is 50–100 IU/kg repeated every
8–12 hours. The response should be assessed clinically and factor VIII levels monitored.
Major bleeding: The recommended dose of factor VIII is 50–150 IU/kg repeated every
8–12 hours. The response should be assessed clinically and factor VIII levels monitored
and maintained at > 50% until healing has completed.
3.2.2. Low titre inhibitor (<5 BU/ml) but history of high responder
Minor bleeding: Infusions of factor VIII will cause an anamnestic rise of the levels of
factor VIII inhibitor within 3–5 days rendering further therapy with factor VIII
ineffective. The recommended adult dose of recombinant factor VIIa is 90 µg/kg
(recommended paediatric dose may be up to 200–250 µg/kg) at 2 hourly intervals for a
minimum of 2 doses or until an objective clinical response is observed. This should be
followed up with a further single dose. If no response is observed treatment with APCC
should be considered.
Major bleeding: The recommended adult dose of recombinant factor VIIa is 90 µg/kg
(recommended paediatric dose may be up to 200–250 µg/kg) at 2 hourly intervals for 12
hours or until an objective clinical response is observed. Dose intervals can be increased
Use of recombinant and plasma-derived Factor VIII and IX
193
to 3 or 4 hours. The duration of therapy depends on the extent of the initial
haemorrhage. If no response is observed, treatment with APCC should be considered.
3.2.3. High titre inhibitor (>5 BU/ml)
Minor bleeding: The recommended adult dose of recombinant factor VIIa is 90 µg/kg
(recommended paediatric dose may be up to 200–250 µg/kg) at 2 hourly intervals for a
minimum of 2 doses or until an objective clinical response is observed. This should be
followed up with a further single dose. If no response is observed treatment with APCC
should be considered.
Major bleeding: The recommended adult dose of recombinant factor VIIa is 90 µg/kg
(recommended paediatric dose may be up to 200–250 µg/kg) at 2 hourly intervals for 12
hours; then increased to 3 hourly intervals depending on the observed clinical response.
If appropriate, the dose interval can be further increased to 4 hours as a maintenance
dosage. The duration of therapy is dependent on the severity of the haemorrhage. If
major surgery has been performed, the duration of therapy may extend to 14 days. An
alternative dosing scheme is 320 µg/kg every 6 hours. If no response is observed,
treatment with APCC should be considered.
3.2.4. Elective Major Surgery
Major surgery in patients with inhibitors carries a high degree of risk and should only be
carried out in recognised HTCs after careful consultation and agreement with at least one
other Australian haemophilia specialist. All such discussions should be documented. It is
recommended that a pharmacokinetic study be undertaken before surgery. The dosage
regimen is based on the regimen for major bleeds (Sections 3.2.1, 3.2.2 and 3.2.3 above).
Dental surgery and the insertion of IV access devices require 3–5 days of therapy and
antifibrinolytics.
3.2.5. Emergency Major Surgery
The dosage regimen is based on the regimen for major bleeds (Sections 3.2.1, 3.2.2 and
3.2.3 above). If time allows, there should be consultation and agreement with one other
haemophilia specialist as in Elective Major Surgery. Any such discussion should be
documented. The patient should be transferred to a recognised HTC as soon as
practicable.
3.2.6. Home Therapy
AHCDO recommends that home therapy only be considered with close monitoring and
regular medical supervision.
Tolerisation
Tolerisation should be considered in all those patients with recent persisting inhibitors.
Written informed consent should be obtained before starting. Intensive replacement
therapy for immune tolerance usually requires central venous access.
194
Use of recombinant and plasma-derived Factor VIII and IX
Haemophilia A
The International Registry on Tolerisation identifies better results in those patients with
lower age at the start of Immune Tolerance Induction (ITI); shorter elapsed time of
inhibitor presence before ITI; lower maximum pre-treatment inhibitor titres and
treatment with higher doses of FVIII.
AHCDO strongly recommends participation in an international randomised trial of high
dose versus low dose factor VIII tolerisation protocols. All patients considered eligible
for the trial should be involved
For patients who do not enter an international trial, there are a number of published
tolerisation protocols that describe a variety of doses of factor VIII (e.g 50 IU/kg three
times a week up to 200 IU/kg daily) as well as the use of immune suppression.
Tolerisation should continue until eradication of the inhibitor demonstrated by a greater
than 60% recovery and normal half-life of factor VIII. Tolerisation should only be
attempted in consultation with a physician experienced in the management of patients
with haemophilia and inhibitors.
Acquired Haemophilia A
Immunosuppression is recommended for eradication of inhibitors in acquired
haemophilia A.
It is recommended that adults with severe haemophilia A who have had an inhibitor for
many years, and in some instances decades, should be managed with treatment of
haemorrhage by infusion of recombinant factor VIIa. Adult patients with mild or
moderate haemophilia A who develop inhibitors often have mild inhibitors, which may
decline with time. These patients should be treated with desmopressin (DDAVP) for
minor bleeds and recombinant factor VIIa for more serious haemorrhages. Tolerisation
may be considered if haemorrhages cannot be controlled by recombinant factor VIIa.
Use of recombinant and plasma-derived Factor VIII and IX
195
Protocols for dental procedures in patients with
haemophilia A, B or von Willebrand disease *
Local
anaesthetic
infiltration
Local
anaesthetic
inferior alveolar
nerve block
Supragingival
scale only
Subgingival scale or a
restorative procedure
requiring a matrix band
Minor soft
tissue
abscess/
swelling
Mild
(>5 IU/dl)
No pretreatment
required
No pre-treatment
required
No pretreatment
required
A pre-treatment dose of 1 g,
oral tranexamic acid capsules
followed by 1 g qid for 24 h
post-treatment
FVIII at 10
IU/kg pretreatment
Moderate
(5–2 IU/dl)
No pretreatment
required
FVIII at 10 IU/kg
pre-treatment
No pretreatment
required
FVIII at 7 IU/kg and a pretreatment dose of 1 g, oral
tranexamic acid capsules
followed by 1 g qid for 3 days
post-treatment
FVIII at 10
IU/kg pretreatment
Severe
(<2 IU/dl)
No pretreatment
required
FVIII at 10 IU/kg
pre-treatment
A pretreatment dose
of 1g only, oral
tranexamic
acid capsules
FVIII at 7 IU/kg and a pretreatment dose of 1g, oral
tranexamic acid capsules
followed by 1 g qid for 3 days
post-treatment
FVIII at 10
IU/kg pretreatment
Mild
No pretreatment
required
No pre-treatment
required
No pretreatment
required
A pre-treatment dose of 1g
oral tranexamic acid capsules
followed by a dose of 1 g quid
for 24 hours post-treatment
ProthrombinexHT at 20 IU/kg
pre-treatment
Moderate
No pretreatment
required
ProthrombinexHT at 20 IU/kg
pre-treatment
No pretreatment
required
Prothrombinex-HT at 14 IU/kg
pre-treatment and a pretreatment dose of 1g oral
tranexamic acid capsules
followed by a dose of 1g qid
for 3 days post-treatment
ProthrombinexHT at 20 IU/kg
pre-treatment
Severe
No pretreatment
required
ProthrombinexHT at 20 IU/kg
pre-treatment
A pretreatment dose
of 1g only oral
tranexamic
acid capsules
Prothrombinex-HT at 14 IU/kg
pre-treatment and a pretreatment dose of 1g oral
tranexamic acid capsules
followed by a dose of 1g qid
for 3 days post-treatment
ProthrombinexHT at 20 IU/kg
pre-treatment
Mild§
(>30 IU/dl)
No pretreatment
required
No pre-treatment
required
No pretreatment
required
A pre-treatment dose of 1 g
oral tranexamic acid capsules
followed by a dose of 1 g qid
for 24 hours post-treatment
Desmopressin
pre-treatment
Moderate§
(10–30
IU/dl)
No pretreatment
required
No pre-treatment
required
No pretreatment
required
A pre-treatment dose of 1 g
oral tranexamic acid capsules
followed by a dose of 1g qid
for 24 hours post-treatment
FVIII at 10
IU/kg or
desmopressin
pre-treatment
Severe§
(<10 IU/dl)
No pretreatment
required
FVIII at 10 IU/kg
pre-treatment
A pretreatment dose
of 1g only
tranexamic
acid capsules
FVIII at 7 IU/kg and a pretreatment dose of 1g oral
tranexamic acid capsules
followed by a dose of 1g qid
for 3 days post-treatment
FVIII at 10
IU/kg pretreatment
Haemophilia A
Haemophilia B
Von Willebrand
disease (Type 1)
*modified from Stubbs and Lloyd 2001; § arbitrary distinctions in disease severity devised by authors; qid = once per day; bd = twice per day;
tds = three times per day
196
Use of recombinant and plasma-derived Factor VIII and IX
Appendix H Critical Appraisal Checklists
Systematic review critical appraisal checklist
Source: Khan et al. 2001
Title of assessment:
Title of systematic review:
Author(s):
Year:
Comparators:
Score :
/6
1.
What is the review’s objective?
What were the population/participants, interventions, outcomes and study designs?
2.
What sources were searched to identify primary studies?
What sources (e.g. databases) were searched and were any restrictions by date,
language and type of publication used? Were other strategies used to identify
research?
3.
What were the inclusion criteria and how were they applied?
4.
What criteria were used to assess the quality of primary studies and how were
they applied?
5.
How were the data extracted from the primary studies?
6.
How were the data synthesised?
How were differences between studies investigated?
How were the data combined? Was it reasonable to combine the studies?
What were the summary results of the review?
Do the conclusions flow from the evidence reviewed?
Use of recombinant and plasma-derived Factor VIII and IX
197
Checklist for appraising the quality of intervention studies
SSST
T
U
D
Y
Q
U
A
L
T
Y
A
E
M
E
N
T
C
H
E
C
K
L
T
TU
UD
DY
YQ
QU
UA
AL
LIIIT
TY
YA
ASSSSSSE
ESSSSSSM
ME
EN
NT
TC
CH
HE
EC
CK
KL
LIIISSST
T
SSSuuuiiitttaaabbbllleee fffooorrr tttrrriiiaaalllsss,,, cccooohhhooorrrtttsss aaannnddd cccaaassseee---cccooonnntttrrrooolll ssstttuuudddiiieeesss aaasssssseeessssssiiinnnggg iiinnnttteeerrrvvveeennntttiiiooonnnsss
(((D
D
w
B
N
B
A
m
Dooow
wnnnsss aaannnddd B
Blllaaaccckkk (((111999999888)))–––aaadddaaapppttteeeddd fffooorrr ttthhhiiisss N
NB
BA
A aaasssssseeessssssm
meeennnttt)))
Clinical practice guidelines for factor VIII and factor IX
Author(s):
Institution(s):
Year:
Study Design:
Comparators:
Possible confounders = age, gender, severity of
the disease, inhibitors, HIV, HAV, HBV,
HCV, previous exposure to factors
Reporting
1. Is the hypothesis/aim/objective of the study
clearly described?
yes
partially
no
Yes
no
2.
Are the main outcomes to be measured clearly
described in the Introduction or Methods
section?
If the main outcomes are first mentioned in the
Results section, the question should be
answered ‘no’.
yes
no
3.
4.
1
0
Are the interventions of interest clearly
described?
Interventions that are to be compared should
be clearly described.
Yes
no
5.
1
0
Are the characteristics of the patients included
in the study clearly described?
In cohort studies and trials, inclusion and/or
exclusion criteria should be given.
Yes
no
6.
1
0
Are the distributions of principal confounders
in each group of subjects to be compared
clearly described?
198
Are the main findings of the study clearly
described?
Simple outcome data (including denominators
and numerators) should be reported for all
major findings so that the reader can check the
major analyses and conclusions (This question
does not cover statistical tests which are
considered below).
Yes
no
7.
2
1
0
1
0
Does the study provide estimates of the
random variability in the data for the main
outcomes?
In non-normally distributed data the interquartile range of results should be reported. In
normally distributed data the standard error,
standard deviation or confidence intervals
should be reported. If the distribution of the
data is not described, it must be assumed that
the estimates used were appropriate and the
question should be answered ‘yes’.
Yes
1
no
0
8. Have all important adverse events that may be
a consequence of the intervention been
reported?
Use of recombinant and plasma-derived Factor VIII and IX
This should be answered ‘yes’ if the study
demonstrates that there was a comprehensive
attempt to measure adverse events.
Adverse events = infections, infectious
diseases, development of inhibitors,
thrombosis, myocardial infarction.
Yes
no
9.
1
0
Have the characteristics of patients lost to
follow-up been described?
This should be answered ‘yes’ where there
were no losses to follow-up or where losses to
follow-up were so small that findings would be
unaffected by their inclusion. This should be
answered ‘no’ where a study does not report
the number of patients lost to follow-up.
Yes
no
1
0
10. Have the actual probability values been
reported (eg 0.035 rather than <0.05) for the
main outcomes, except where the probability
value is less than 0.001?
yes
no
1
0
External validity
All the following criteria attempt to address the
representativeness of the findings of the study and
whether they may be generalised to the population
from which the study subjects were derived.
11. Were the subjects asked to participate in the
study representative of the entire population
from which they were recruited?
The study must identify the source population
for patients and describe how the patients were
selected. Patients would be representative if
they comprised the entire source population, an
unselected sample of consecutive patients, or a
random sample. Random sampling is only
feasible where a list of all members of the
relevant population exists. Where a study does
not report the proportion of the source
population from which the patients are derived,
the question should be answered as ‘unable to
determine’.
Yes
1
no
0
unable to determine 0
12. Were those subjects who were prepared to
participate representative of the entire
population from which they were recruited?
The proportion of those asked who agreed
should be stated. Validation that the sample
was representative would include
demonstrating that the distribution of the main
confounding factors was the same in the study
sample and the source population.
Yes
no
unable to determine
1
0
0
13. Were the staff, places, and facilities where the
patients were treated, representative of the
treatment the majority of patients receive?
For the question to be answered ‘yes’ the study
should demonstrate that the intervention was
representative of that in use in the source
population. The question should be answered
‘no’ if, for example, the intervention was
undertaken in a specialist centre
unrepresentative of the hospitals most of the
source population would attend.
Yes
no
unable to determine
1
0
0
Internal validity – Bias
14. Was an attempt made to blind study subjects to
the intervention they have received?
For studies where the patients would have no
way of knowing which intervention they
received, this should be answered ‘yes’.
Yes
no
unable to determine
1
0
0
15. Was an attempt made to blind those measuring
the main outcomes of the intervention?
Yes
no
unable to determine
1
0
0
16. If any of the results of the study were based on
‘data dredging’, was this made clear?
Any analyses that had not been planned at the
outset of the study should be clearly indicated.
If no retrospective unplanned subgroup
analyses were reported, then answer ‘yes’.
Yes
1
no
0
unable to determine 0
17. In trials and cohort studies, do the analyses
adjust for different lengths of follow-up of
patients?
Where follow-up was the same for all study
patients the answer should be ‘yes’. If different
Use of recombinant and plasma-derived Factor VIII and IX
199
lengths of follow-up were adjusted for by, for
example, survival analysis the answer should
be ‘yes’. Studies where differences in followup are ignored should be answered ‘no’.
yes
no
unable to determine
1
0
0
18. Were the statistical tests used to assess the
main outcomes appropriate?
The statistical techniques used must be
appropriate to the data. For example nonparametric methods should be used for small
sample sizes. Where little statistical analysis
has been undertaken but where there is no
evidence of bias, the question should be
answered ‘yes’. If the distribution of the data
(normal or not) is not described it must be
assumed that the estimates used were
appropriate and the question should be
answered ‘yes’.
Yes
no
unable to determine
1
0
0
19. Was compliance with the intervention/s
reliable?
Where there was non-compliance with the
allocated treatment or where there was
contamination of one group, the question
should be answered ‘no’. For studies where the
effect of any misclassification was likely to
bias any association to the null, the question
should be answered ‘yes’.
Yes
no
unable to determine
1
0
0
20. Were the main outcome measures used
accurate (valid and reliable)?
For studies where the outcome measures are
clearly described, the question should be
answered ‘yes’. For studies which refer to
other work or that demonstrates the outcome
measures are accurate, the question should be
answered ‘yes’.
Yes
no
unable to determine
1
0
0
Internal validity – Confounding (selection bias)
21. Were the patients in different intervention
groups (trials and cohort studies) recruited
from the same population?
200
For example, patients for all comparison
groups should be selected from the same
hospital. The question should be answered
‘unable to determine’ where there is no
information concerning the source of patients
included in the study.
Yes
no
unable to determine
1
0
0
22. Were study subjects in different intervention
groups (trials and cohort studies) recruited
over the same period of time?
For a study which does not specify the time
period over which the patients were recruited,
the question should be answered as ‘unable to
determine’.
Yes
no
unable to determine
1
0
0
23. Were study subjects randomised to
intervention groups?
Studies which state that subjects were
randomised should be answered ‘yes’ except
where method of randomisation is unknown or
would not ensure random allocation. For
example, alternate allocation would score ‘no’
because it is predictable.
Yes
no
unable to determine
1
0
0
24. Was the randomised intervention assignment
concealed from both patients and health care
staff until recruitment was complete and
irrevocable?
All non-randomised studies should be
answered ‘no’. If assignment was concealed
from patients but not from staff, it should be
answered ‘no’.
yes
no
unable to determine
1
0
0
25. Was there adequate adjustment for
confounding in the analyses from which the
main findings were drawn?
This question should be answered ‘no’ for
trials if: the main conclusions of the study
were based on analyses of treatment rather
than intention-to-treat; the distribution of
known confounders in the different treatment
groups was not described; or the distribution of
known confounders differed between the
Use of recombinant and plasma-derived Factor VIII and IX
treatment groups but was not taken into
account in the analyses. In non-randomised
studies if the effect of the main confounders
was not investigated or confounding was
demonstrated but no adjustment was made in
the final analyses the question should be
answered as ‘no’.
yes
no
unable to determine
1
0
0
26. Were losses of patients to follow-up taken into
account?
If the number of patients lost to follow-up are
not reported, the question should be answered
as ‘unable to determine’. If the proportion lost
to follow-up was too small to affect the main
findings, the question should be answered
‘yes’.
Yes
no
unable to determine
1
0
0
Subscale Scores
Reporting =
External validity =
Bias =
Confounding =
/11
/3
/7
/6
Total Quality Index Score = /27
Power
27. Did the study have sufficient power to detect a
clinically important effect where the
probability value for a difference being due to
chance is less than 5%?
a. Was there enough power to detect a
difference of ..%, in the outcome ‘….’?
Sample sizes – n1 = n2 =
power =
Use of recombinant and plasma-derived Factor VIII and IX
201
Checklist for the critical appraisal of case series
Source: Young et al. 1999, Lung volume reduction surgery (LVRS) for chronic obstructive pulmonary disease
(COPD) with underlying severe emphysema. A West Midlands Development and Evaluation
Committee Report, University of Birmingham, pp. 51–53.
Title of review:
Title of study:
Author(s):
Year:
Comparators:
Score:
1.
/3
Was the study conducted prospectively?
Were the key outcomes measured before and after the intervention, using clear
criteria defined a priori?
2. Was the method of selection of cases identified and appropriate?
Were patients selected consecutively or in an unbiased manner?
Was there evidence that the characteristics of the included cases were not
significantly different from those of the treated population?
3.
Was the duration and completeness of follow-up reported and was it
adequate?
Are the number and characteristics of losses to follow-up presented? #
Are losses to follow-up managed by performing sensitivity analysis and/or including
them in the final analysis?
#
Losses to follow-up >20% are unacceptable, particularly if unaccounted for.
202
Use of recombinant and plasma-derived Factor VIII and IX
Checklist for assessing the clinical importance of benefit or
harm
Rank scoring for appraising the clinical importance of benefit/harm
Source: NHMRC 2000b
Title of review:
Title of study:
Author(s):
Year:
Comparators:
Clinically important effect:
Rank Score :
Ranking
1
/4
Clinical importance of benefit/harm
A clinically important benefit for the full range of plausible estimates.
The confidence limit closest to the measure of no effect (the ‘null’) rules out a clinically unimportant
effect of the intervention.
2
The point estimate of effect is clinically important but the confidence interval includes clinically
unimportant effects.
3
The confidence interval does not include any clinically important effects.
4
The range of estimates defined by the confidence interval includes clinically important effects
but the range of estimates defined by the confidence interval is also compatible with no effect,
or a harmful effect.
Use of recombinant and plasma-derived Factor VIII and IX
203
Checklist for assessing the relevance of outcomes
Rank scoring for classifying the relevance of evidence
Source: NHMRC 2000b
Title of review:
Title of study:
Author(s):
Year:
Comparators:
Rank Score :
/5
Ranking
Relevance of the evidence
1
Evidence of an effect on patient-relevant clinical outcomes, including benefits and harms, and quality of
life and survival.
2
Evidence of an effect on a surrogate outcome that has been shown to be predictive of patient-relevant
outcomes for the same intervention.
3
Evidence of an effect on proven surrogate outcomes but for a different intervention.
4
Evidence of an effect on proven surrogate outcomes but for a different intervention and population.
5
Evidence confined to unproven surrogate outcomes.
204
Use of recombinant and plasma-derived Factor VIII and IX
Appendix I
Evidence Tables
Summary tables for intervention studies
(NHMRC Guidelines Assessment Register Consultants Working Party 2004)
STUDY DETAILS
Reference [1] (Arkin et al 1991)
Affiliation/source of funds [2] rFVIII Clinical Trial Group, partially funded by Cutter Biological, Miles Inc.
Study design [3] historical control study
Level of evidence [4] III-3
Intervention [6] rFVIII prophylaxis therapy
Location/setting [5] Home care setting,
worldwide centres
Comparator(s) [8] pdFVIII (Koate HS) treatment
(20-40 IU/kg, 3 x week)
Sample size [7]14 (Stage I)
Sample size [9] Not stated
Patient characteristics [10]
Intervention group – moderate-severe haemophilia A patients aged 4-72 without inhibitors, PTPs
Comparator group(s) – no info provided on historical control group
Length of follow-up [11] Not reported
Outcome(s) measured [12] In vivo recovery FVIII, adverse events
INTERNAL VALIDITY
Allocation [13] Historical
control
Comparison of study groups
[14] Unable to ascertain
Blinding [15] No
Treatment/ measurement bias
[16] Possible
Follow-up (ITT) [17]
Unable to ascertain
Overall quality assessment (descriptive) [18] Quality score: 5/27
Selection bias likely
Confounding likely
Follow-up uncertain
External validity unclear
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Incremental FVIII recovery
2.7±0.6
2.5±0.33
Measure of effect/effect
size [22]
Benefits (NNT) [23]
8.0% difference in
recovery
p=0.29
95% CI [25]
Harms (NNH) [24]
95% CI [25]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27] 2
Any other adverse effects [28] ‘uncommon and mild’- erythema, cold hands/feet, burning at infusion site, dry mouth. More severe= dizziness,
hypotension, rash, shortness of breath. Frequency of adverse reactions= 1%
EXTERNAL VALIDITY
Generalisability [29] Unclear
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
205
STUDY DETAILS
Reference [1] (Cattaneo et al 1989)
Affiliation/source of funds [2] National Institute of Health grant
Study design [3] Randomised controlled crossover
trial
Level of evidence [4] II
Location/setting [5]
1 treatment centre, Milan, Italy
Intervention [6]
Comparator(s) [8]
Cryoprecipitate + DDAVP (0.3µg/kg)
Cryoprecipitate + saline
Sample size [7] 10
Sample size [9] 10
Patient characteristics [10]
10 PTPs with severe vWD, (type not specified) with prolonged bleeding time (>30 minutes) with need for cryoprecipitate for joint or muscle
haemorrhage
Length of follow-up [11] Crossover at day 15, follow-up not
stated
Outcome(s) measured [12] BT, vWF:Ag, vWF:RiCof
INTERNAL VALIDITY
Allocation [13] random
(method not specified)
Comparison of study groups
[14] same patients –
crossover
Blinding [15]
Double- both
patient and
operator
Treatment/ measurement bias
[16] same patients – crossover
Follow-up (ITT) [17]
ITT used
complete
Overall quality assessment (descriptive) [18] Unable to assess external validity
Bias minimised
Confounding avoided
RESULTS
Outcome [19]
No significant difference
between cryo and saline,
but significant between
cryo and DDAVP.
Intervention group [20]
BT After cryo=12±2
minutes
Control group [21]
BT after cryo= 15±3
minutes
Measure of effect/effect
size [22]
Benefits (NNT) [23]
P<0.01
BT After DDAVP=9±2
minutes
BT after saline=
15±3
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 1/4 Clinically important difference
in BT
Relevance (1-5) [27] 1/5
Any other adverse effects [28] Not reported
EXTERNAL VALIDITY
Generalisability [29] Only severe patients (type not specified)
Applicability [30]
Comments [31]
206
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (De Sio et al 1985)
Affiliation/source of funds [2] Not stated
Study design [3] non randomised controlled trial
Level of evidence [4] III-2
Location/setting [5] 1 treatment centre,
Rome, Italy
Comparator(s) [8] Subcutaneous DDAVP (0.3µg/kg)
Intervention [6] Intravenous DDAVP (0.3µg/kg)
Sample size [9] 16
Sample size [7] 18
8 people received both
Patient characteristics [10] 26 patients, 21 mild haemophilia A, 5 moderate haemophilia A
Intervention group –13 mild haemophilia A, 5 moderate haemophilia A
Comparator group(s) –13 mild haemophilia A, 3 moderate haemophilia A
Length of follow-up [11] 2 hours
Outcome(s) measured [12] FVIII:C ratio post/pre
INTERNAL VALIDITY
Allocation [13] Not stated
Comparison of study groups
[14] Not stated
Blinding [15] No
Treatment/ measurement bias
[16] Unclear
Follow-up (ITT) [17]
Adequate
Overall quality assessment (descriptive) [18]
Bias possible
Confounding possible
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
No statistically significant
differences between
treatments in FVIII:C
activity at 60 minutes post
DDAVP
I hour post
I hour post
FVIII:C Mean=24.9
FVIII:C Mean=25.4
SD=12.4
SD=12.4
Measure of effect/effect
size [22]
Benefits (NNT) [23]
t=.288, p>0.5
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 3/4
Relevance (1-5) [27] None
Any other adverse effects [28] Moderate increase of pulse rate and/or blood pressure, frequently associated with mild facial flushing.
EXTERNAL VALIDITY
Generalisability [29] Unable to assess external validity
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
207
STUDY DETAILS
Reference [1] (Ewenstein et al 2002)
Affiliation/source of funds [2] Mononine Comparison Study Group. (Funded in part by MonoFIX (Aventis Behring, King of Prussia, PA)
Study design [3] Randomised crossover study
Level of evidence [4] II
Location/setting [5]
15 haemophilia centres in United States
Intervention [6] Single bolus infusion (50 IU/kg) – non bleeding
Comparator(s) [8] Single bolus infusion (50 IU/kg) – non bleeding
rFIX- 7 day washout
pdFIX- 7 day washout
Sample size [7] 38 (crossover design)
Sample size [9] 38 (crossover design)
Patient characteristics [10] Clinically diagnosed with moderate-severe haemophilia B, aged > 5 years, prior treatment with FIX. No patients had
inhibitors, acquired haemophilia, abnormal prothrombin time
Intervention group and Comparator group – median age 18.5 (range 7-75)
FIX:C <1%= 14
FIX:C 1-5%=21
FIX:C >5%=3
Length of follow-up [11] 48 hours
Outcome(s) measured [12] recovery of pd/rFIX -> plasma level FIX:C (IU/dL)
INTERNAL VALIDITY
Allocation [13]
randomisation protocol
not stated
Comparison of study groups
[14] same participants
Blinding [15]
double-blind
Treatment/ measurement bias
[16] none
Follow-up (ITT) [17]
Done
Overall quality assessment (descriptive) [18]
Selection bias minimised; confounding avoided; patients representative; follow-up adequate
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
recovery FIX (IU d/L rise
per IU/kg infused)
0.86±0.31 IU/dL/kg
1.71±0.73 IU/dL/kg
Measure of effect/effect
size [22]
Benefits (NNT) [23]
0.85 IU/dL/IU/Kg
95% CI [25]
95% CI [25] [0.66-1.04]
p<0.0001
Clinical importance (1-4) [26] 1/4
Harms (NNH) [24]
95% CI [25]
Relevance (1-5) [27] 2/5
Any other adverse effects [28] Not reported
EXTERNAL VALIDITY
Generalisability [29] PTPs in non-bleeding state
Applicability [30]
Comments [31]
208
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Fijnvandraat et al 1997; Berntorp 1997)
Affiliation/source of funds [2]Pharmacia AB
Study design [3] Randomised single-blinded
crossover trial
Level of evidence [4] II
Location/setting [5] Multicentre, Sweden and
the Netherlands
Intervention [6] rFVIII SQ
(50 IU/kg)
Comparator(s) [8] pdFVIII
(50 IU/kg, Octonativ)
Sample size [7] 12
Sample size [9] 12
Patient characteristics [10] 12 PTPs with severe haemophilia A, aged 17-64 years, without inhibitors
Intervention group – as above
Comparator group(s) –as above
Length of follow-up [11] 50 hours
Outcome(s) measured [12] Incremental FVIII recovery (IU/kg-1) % change
INTERNAL VALIDITY
Allocation [13]
randomised
Comparison of study groups
[14] same patients
Blinding [15]
single blinded
(patient)
Treatment/ measurement bias
[16] possible
Follow-up (ITT) [17]
complete
Overall quality assessment (descriptive) [18]
Selection bias minimised; confounding avoided; follow-up adequate; patients representative
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Incremental FVIII recovery
(IU/kg-1)
2.4±0.21
2.4±0.26
Measure of effect/effect
size [22] percentage
change=0
95% CI [25]
Benefits (NNT) [23]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27]
Any other adverse effects [28] None reported
EXTERNAL VALIDITY
Generalisability [29]
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
209
STUDY DETAILS
Reference [1] (Giles et al 1998)
Affiliation/source of funds [2] Association of Hemophilia Centre Directors of Canada (AHCDC)- partial funding/products provided by Bayer Corpnot clear
Study design [3] non randomised controlled trail
Level of evidence [4] III-2
Location/setting [5] Haemophilia Centres in
Canada
Intervention [6] PTPs transferred to rFVIII
Comparator(s) [8] PTPs transferred to affinity-purified FVIII
Sample size [7] 814
Sample size [9] 58
Patient characteristics [10]
Intervention group – haemophilia A participants switched to rFVIII
Comparator group(s) –haemophilia A participants switched to affinity purified FVIII
Length of follow-up [11] 2 years
Outcome(s) measured [12] level of FVIII inhibitors (Bethesda assay)
INTERNAL VALIDITY
Allocation [13] patient
preference
Comparison of study groups
[14] similar inhibitors at
baseline
Blinding [15] Not
done
Treatment/ measurement bias
[16] random variation in followup across participants
Follow-up (ITT) [17]
<60%
Overall quality assessment (descriptive) [18] relevant to real situation but high loss to follow-up
Selection bias not minimised (patient self-selection)
Confounding unclear
Follow-up adequate
External validity uncertain
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Bethesda assay
Year 1
Year 2
Year 1
Year 2
0
74.3%
79.7%
75.4%
78.1%
<0.5
22.0%
15.3%
17.5%
15.6%
>0.5
3.8%
5%
7.0%
6.3%
Measure of effect/effect
size [22]
Benefits (NNT) [23]
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27]
Any other adverse effects [28] None reported
EXTERNAL VALIDITY
Generalisability [29] Participants are good representation
Applicability [30]
Comments [31]
210
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Kadir et al 2002)
Affiliation/source of funds [2] Ferring Pharmaceuticals, Katherine Dormandy Trust
Study design [3] randomised controlled crossover
trial
Level of evidence [4] II
Location/setting [5]
Home treatment, United Kingdom
Intervention [6]
Comparator(s) [8]
DDAVP (300µg) intranasal
Placebo nasal spray
Sample size [9]
Sample size [7]
Patient characteristics [10] 39 women with menorrhagia. Mild-moderate vWD, heterozygote FXI-deficiency, and carriers of haemophilia
(DDAVP/placebo)- mean age= 32.2 years Baseline Mean PBAC score =292.1
(placebo/DDAVP)- mean age=31.6 years Baseline Mean PBAC score=262.1
Length of follow-up [11] 2 months
Outcome(s) measured [12] Mean difference in PBAC (Pictorial Blood
Assessment Chart) scores
Side effects
INTERNAL VALIDITY
Allocation [13]
randomised (method not
specified)
Comparison of study groups
[14] No significant
differences between
Blinding [15]
double-blinded
Treatment/ measurement bias
[16] Treated and measured the
same
Follow-up (ITT) [17]
Did use ITT, except
those who did not
return diaries.
Overall quality assessment (descriptive) [18] Quality score: 24/27
Unable to assess external validity
Bias minimised
Confounding avoided
RESULTS
Outcome [19]
Both placebo and DDAVP
resulted in significant
decrease in PBAC score
from baseline.
Insignificant different
between placebo and
DDAVP
Intervention group [20]
PBAC score- 140
Control group [21]
PBAC score- 148
Measure of effect/effect
size [22]
Benefits (NNT) [23]
No significant differences
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27]
Any other adverse effects [28] 83 % experienced one or more adverse events. 69% on DDAVP, 52% on placebo (p=0.3)
Headache, weight gain
EXTERNAL VALIDITY
Generalisability [29] Patients representative
Applicability [30] More benefit than harm
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
211
STUDY DETAILS
Reference [1] (Kelly et al 1997)
Affiliation/source of funds [2] Products supplied by Baxter-Hyland corporation
Division of Haematology, Children’s hospital of Philadelphia, University of Pennsylvania, Philadelphia
Study design [3] quasi- randomised controlled trial
Level of evidence [4] III-1
Location/setting [5] Children’s hospitalUnited States
Intervention [6] Recovery study in non-bleeders
Comparator(s) [8]
Hemofil M (50 IU/kg) -> 72 hours-> Recombinate (50 IU/kg)
Hemofil M (50 IU/kg) -> 72 hours-> Hemofil M
Sample size [7] 10
Sample size [9] 10
Patient characteristics [10] 10 haemophilia A patients aged 7-12 years without inhibitors, PTPs
Intervention group – 2nd 5 participants
Comparator group(s) –1st 5 participants
Length of follow-up [11] 72 hours
Outcome(s) measured [12] Recovery/response rate, inhibitor (Bethesda
assay)
INTERNAL VALIDITY
Allocation [13] sequential
5 participants
Comparison of study groups
[14] same participants
Blinding [15]
double-blind
Treatment/ measurement bias
[16] same participants
Follow-up (ITT) [17]
complete
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding avoided
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Response
Intervention group [20]
Control group [21]
Recombinate
Hemofil
1.91±0.14%
1.5±0.15%
(p=0.007)
Recovery
Ratio actual: predicted
recovery
100.5±4.5%
78.7±6.2%
(p=0.007)
>0.66 in 100%
patients
<0.66 in 30%
patients
Measure of effect/effect
size [22]
Benefits (NNT) [23]
95% CI [25]
95% CI [25]
Clinical importance (1-4) [26] 2/4
Harms (NNH) [24]
95% CI [25]
Relevance (1-5) [27] 2/5
Any other adverse effects [28] none reported
EXTERNAL VALIDITY
Generalisability [29] Participants are representative of paediatric patients (PTPs)
Applicability [30] Potential benefits of rFVIII outweigh potential harms
Comments [31] Significantly better response and recovery with rFVIII compared to pdFVIII
212
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Kisker et al 2003)
Affiliation/source of funds [2] The Mononine Study group, financially supported by Aventis Behring
Study design [3] randomised crossover trial
Level of evidence [4] II
Location/setting [5] University of Iowa, USA
Intervention [6] rFIX
(50 IU/kg, BeneFIX®)
Comparator(s) [8] pdFIX
(50 IU/kg, MonoFIX®)
Sample size [7] 15
Sample size [9] 15
Patient characteristics [10] 15 non-bleeding severe haemophilia B patients, aged >12 years, without inhibitors
Intervention group – as above
Comparator group(s) –as above
Length of follow-up [11] 7-30 days
Outcome(s) measured [12] Incremental factor recovery, IU/kg-1, % change
INTERNAL VALIDITY
Allocation [13] random –
not stated how
Comparison of study groups
[14] same patients
Blinding [15]
double blinded
Treatment/ measurement bias
[16] minimised
Follow-up (ITT) [17]
complete
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding avoided
Follow-up adequate
Good external validity
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Incremental factor
recovery, IU/kg-1
0.86
(0.39-1.48)
1.67
(0.86-4.66)
Measure of effect/effect
size [22]
Benefits (NNT) [23]
48.5%, p=0.002
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 1/4
Relevance (1-5) [27] 2/5
Any other adverse effects [28] Not reported. Follow-up probably too short for development of inhibitors
EXTERNAL VALIDITY
Generalisability [29] Representative
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
213
STUDY DETAILS
Reference [1] (Kreuz et al 2002)
Affiliation/source of funds [2]
Study design [3] Historical control group
Level of evidence [4] III-3
Location/setting [5] Haemophilia centre,
University Hospital, Frankfurt am Main,
Germany
Intervention [6]
Comparator(s) [8]
rFVIII treatment: on-demand, prophylaxis, surgery
pdFVIII treatment: on-demand, prophylaxis, surgery
Sample size [7] 21
Sample size [9] 51
Patient characteristics [10] PUPs with moderate-severe haemophilia A, no detectable inhibitors
Intervention group – participants after 1989 treated with rFVIII
Comparator group(s) –participants up to 1989 treated with pdFVIII
Length of follow-up [11] 23 years
Outcome(s) measured [12] inhibitor development
INTERNAL VALIDITY
Allocation [13] Historical
Comparison of study groups
[14] Unsure
Blinding [15] None
Treatment/ measurement bias
[16] Possible
Follow-up (ITT) [17]
complete
Overall quality assessment (descriptive) [18]
Selection bias likely
Confounding possible
Follow-up unclear
Patients representative
RESULTS
Outcome [19]
Inhibitor development
Intervention group [20]
Control group [21]
4 (19.0%) high titre
13 (25.5%) high titre
0 low titre
5 (9.8%) low titre
Measure of effect/effect
size [22]
95% CI [25]
95% CI [25]
Exposure days prior to
inhibitor development
22 (31%)
Benefits (NNT) [23]
Harms (NNH) [24]
15 (4-195)
95% CI [25]
Clinical importance (1-4) [26] None
Relevance (1-5) [27]
Any other adverse effects [28] none reported
EXTERNAL VALIDITY
Generalisability [29] Representative population- long term follow-up, despite this- small sample size
Applicability [30]
Comments [31]
214
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference (Mannucci et al 1987)
Affiliation/source of funds [2] Supported by grants from Ferring AB and Valeas
Study design [3] randomised crossover study
Level of evidence [4] II
Location/setting [5] Malmö, Sweden
Intervention [6] Intravenous DDAVP (0.3µg/kg)
Comparator(s) [8] Subcutaneous DDAVP (0.3µg/kg)
Sample size [7] 14
Sample size [9] 14
Patient characteristics [10] mild and moderate haemophilia A patients (median FVIII:C level 7 U/dL, range 2-31)
Intervention group –as above
Comparator group(s) –as above
Length of follow-up [11] 24 hours- recovery study
Outcome(s) measured [12] pharmacokinetics – peak and half-life FVIII:C
INTERNAL VALIDITY
Allocation [13]
randomised crossover
Comparison of study groups
[14] same participants
Blinding [15]
Treatment/ measurement bias
[16] same participants, bias
minimised
Follow-up (ITT) [17]
ITT
done
Overall quality assessment (descriptive) [18]
Unable to assess external validity
Bias minimised
Confounding avoided
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Measure of effect/effect
size [22]
Benefits (NNT) [23]
Peak FVIII:C mean±/sd
2363±2368
582±158
p<0.02
95% CI [25]
Half-life FVIII:C
4.4±1.1
4.7±1.6
NS
95% CI [25]
Clinical importance (1-4) [26]
Harms (NNH) [24]
95% CI [25]
Relevance (1-5) [27]
Any other adverse effects [28] None reported
EXTERNAL VALIDITY
Generalisability [29] unclear
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
215
STUDY DETAILS
Reference [1] (Mannucci et al 2002)
Affiliation/source of funds [2]
Grant from Alpha therapeutics
Study design [3]
Level of evidence [4] II
Location/setting [5]
Randomised crossover trial
Multicentre study
Comparator(s) [8] Alphanate® Solvent Detergent/Heat
Treated (A-SD/HT)
Intervention [6] Alphanate® Solvent Detergent (A-SD)
60 vWF:Rcof IU/kg
60 vWF:Rcof IU/kg
Sample size [7] 6 then crossover with 6
Sample size [9] 6 then crossover with 6
Patient characteristics:
12 patients with type 3 von Willebrand disease
Length of follow-up [11]
Outcome(s) measured [12] Bleeding time, in vivo half lives of
FVIII:C, vWF:Ag, vWF:Rcof, number of bleeding episodes
186 days
INTERNAL VALIDITY Quality score: 22/27
Allocation [13]
Randomised (not
specified how)
Comparison of study
groups [14] No sig
diffs
Blinding [15]
Not stated
Treatment/
measurement bias
[16] Minimised
Follow-up (ITT) [17]
Follow-up adequate. Not ITT as
one Pp was found to have
inhibitors and was excluded from
analysis. Data given.
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding minimised
Follow-up adequate
External validity unclear
RESULTS
Outcome [19]
A-SD [20]
A-SD/HT [21]
Median bleeding time at
baseline over 30 minutes.
At 1 hour post infusion
was 10.5 minutes.
In vivo half life
FVIII:C= 20.9
vWF:Ag=12.4
vWF:Rcof=7.1
In vivo half life
FVIII:C= 23.8
vWF=12.9
vWF:Rcof=6.5
Measure of effect/effect
size [22]
Benefits (NNT)
[23]
No sig diff between
treatments
95% CI [25]
95% CI [25]
Harms (NNH)
[24]
95% CI [25]
Clinical importance (1-4) [26] 3/4
Relevance (1-5) [27]
Any other adverse effects [28] none
EXTERNAL VALIDITY
Generalisability [29] Unable to determine
Applicability [30] The benefits outweigh any harms
Comments [31] Not available in Australia. Lower purity than recombinant, so not for HA. For use in vWD patients.
216
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Morfini et al 1992)
Affiliation/source of funds [2] Baxter Healthcare Corporation
Study design [3] non randomised crossover trial
Level of evidence [4] III-2
Location/setting [5] Multicentre, Europe and
United States
Intervention [6] All had rFVIII
(50 IU/kg, RecombinateTM) one week after Hemofil
Comparator(s) [8] All patients had pdFVIII
(50 IU/kg, Hemofil®)
Sample size [7] 47
Sample size [9] 47
Patient characteristics [10] 47 PTPs with severe haemophilia A, without inhibitors, same for both intervention and comparators
Length of follow-up [11] 24 hours
Outcome(s) measured [12] Incremental FVIII recovery (IU/kg-1) % change, In
vivo recovery (%)
INTERNAL VALIDITY
Allocation [13] All had
pdfVIII followed by rFVIII
one week later
Comparison of study groups
[14] Same
Blinding [15] Not
stated
Treatment/ measurement bias
[16] Unlikely
Follow-up (ITT) [17]
Complete
Overall quality assessment (descriptive) [18] Quality score: 16/27
Selection bias possible
Confounding avoided
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Measure of effect/effect
size [22]
Benefits (NNT) [23]
Incremental FVIII recovery
2.31
1.99
16.1% difference in
recovery
95% CI [25]
107.1±45.6
92.2±32.0
14.9% difference in in vivo
recovery
In vivo recovery (%)
p<0.019
Harms (NNH) [24]
95% CI [25]
95% CI [25]
Clinical importance (1-4) [26] 3/4
Relevance (1-5) [27] 2/5
Any other adverse effects [28] None reported
EXTERNAL VALIDITY
Generalisability [29] Representative
Applicability [30] More benefits than harms
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
217
STUDY DETAILS
Reference [1] (Poon et al 2002)
Affiliation/source of funds [2] Association of Hemophilia Clinic Directors of Canada (AHCDC) &* Canadian Association of Nurses in Hemophilia
Care
Study design [3] historical control study
Level of evidence [4] III-3
Location/setting [5] Hemophilia Clinics (16)
in Canada
Intervention [6] rFIX 50 IU/kg
Comparator(s) [8] pdFIX (prior infusions)
Sample size [7] 126
Sample size [9] 78
Patient characteristics [10] haemophilia B patients aged 1-74
Severe
Moderate
Mild
Intervention group –
61 (48.4%)
24 (19%)
41 (32.5%)
Comparator group(s) –
44 (59.5%)
12 (16.2%)
18 (24.3%)
Length of follow-up [11] up to 5 years
Outcome(s) measured [12] recovery of FIX -> (expected rise in FIX levels of
50 U/dL)
INTERNAL VALIDITY
Allocation [13] N/A
Comparison of study groups
[14] N/A
Blinding [15] N/A
Treatment/ measurement bias
[16] N/A
Follow-up (ITT) [17]
Overall quality assessment (descriptive) [18] Good
Selection bias uncertain
Confounding possible
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Incremental FIX recovery,
% IU/kg-1 (50 IU/kg)
0.77±0.19
1.05±0.26
Measure of effect/effect
size [22]
Benefits (NNT) [23]
% change= 26.7%
p-value <0.0001
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 1/4
Relevance (1-5) [27] 2/5
Any other adverse effects [28] Anaphylactic reaction
From Canadian registry data: 2 patients (of 244) exposed to rFIX for 1-5 years developed anaphylactic reactions
EXTERNAL VALIDITY
Generalisability [29] Representative population
Applicability [30] Potential benefits outweigh potential harms
Comments [31] Lower recovery rate is based on 1:1 ratio. Current recommendations are 1:1.25 – This study suggests using 1.29x more
recombinant than plasma-derived FIX
218
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Schiavoni et al 1983)
Affiliation/source of funds [2]
Not stated
Study design [3]
Level of evidence [4]
Location/setting [5]
Randomised controlled trail, double blinded
II
Haemophilia summer camp, Sweden
Intervention [6]
Comparator(s) [8]
Tranexamic acid 25mg/kg 3x daily as prophylaxis
Placebo
Sample size [7] 26 in total. Not stated how many in each group.
Patient characteristics [10]
26 patients: 17 severe haemophilia A, 2 mild haemophilia A, 7 severe haemophilia B- Not stated what characteristics of treatment groups were.
Length of follow-up [11] 2 weeks
Outcome(s) measured [12] Number of spontaneous and traumatic bleeding
episodes, amount of FVIII used.
INTERNAL VALIDITY
Allocation [13]
Randomised- method
unknown
Comparison of study groups
[14] Unknown
Blinding [15]
double (unclear
who)
Treatment/ measurement bias
[16] unlikely
Follow-up (ITT) [17]
Adequate
Overall quality assessment (descriptive) [18]
External validity low
Bias minimised
Confounding avoided
Reporting poor
RESULTS
Outcome [19]
Intervention group
[20] 11 traumatic
bleeds
6 spontaneous
bleeds
589 units FVIII used
per bleeding episode
533 units FIX used
per bleeding episode
Control group [21] 14
traumatic bleeds
Measure of effect/effect
size [22]
Benefits (NNT) [23]
4 spontaneous
bleeds
No statistically significant
differences between
groups due to small
sample sizes
95% CI [25]
95% CI [25]
95% CI [25]
825 units FVIII used
per bleeding episode
637 units FIX used
per bleeding episode
Clinical importance (1-4) [26] 3/4
Harms (NNH) [24]
Relevance (1-5) [27]
Any other adverse effects [28] none reported
EXTERNAL VALIDITY
Generalisability [29] Unable to determine external validity.
Applicability [30] Not clear
Comments [31] Prophylaxis with rFVIII or rFIX should now be used in most of this population, therefore benefit of tranexamic acid for
prophylaxis is questionable.
Use of recombinant and plasma-derived Factor VIII and IX
219
STUDY DETAILS
Reference [1] (Seremetis et al 1999) (Schwartz et al 1990)
Affiliation/source of funds [2] The Recombinant FVIII Study Group
Study design [3] non randomised crossover study
Level of evidence [4] III-2
Location/setting [5] Multicentre-worldwide
Intervention [6]
Comparator(s) [8]
rFVIII (Cutter Biological)
PdFVIII (Koate-HS)
1 x IV single bolus- non bleeding Stage 1.
1 x IV single bolus
Sample size [7] 17
Sample size [9] 17
Patient characteristics [10] 16 severe, 1 moderate haemophilia A, PTPs, without inhibitors
Intervention group – as above
Comparator group(s) –as above
Length of follow-up [11] 48 hours- recover study
Outcome(s) measured [12] Recovery, aPTT, length of treatment
INTERNAL VALIDITY
Allocation [13] no
randomised crossover
Comparison of study groups
[14] same participants
Blinding [15] none
Treatment/ measurement bias
[16] same participants
Follow-up (ITT) [17]
done
Overall quality assessment (descriptive) [18]
Selection bias unclear
Confounding avoided
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Intervention group [20]
Mean incremental
recovery
Week 1= 2.68±0.52
(p=0.026)
Week 13= 2.7±0.61
(p=0.20)
Control group [21]
pd-FVIII
Measure of effect/effect
size [22]
2.42±0.33% per
IU/kg
95% CI [25]
Benefits (NNT) [23]
95% CI [25]
Week 25= 2.92±0.99
(p=0.017)
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 3/4
Relevance (1-5) [27]
Any other adverse effects [28] mild-moderate adverse events associated with Kogenate®:
Burning/erythema at infusion site, nausea, pruritis, dizziness, rash, dyspnea, chest tightness, dry mouth; no inhibitors detected
EXTERNAL VALIDITY
Generalisability [29]
Applicability [30]
Comments [31]
220
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Soucie et al 2000)
Affiliation/source of funds [2] Hemophilia Surveillance System
Study design [3] Retrospective cohort
study
Level of evidence [4] III-2
Location/setting [5] information from physicians, clinical laboratories,
hospitals, and haemophilia treatment centres in Colorado, Georgia,
Louisiana, Massachusetts, New York and Oklahoma, United States
Comparator(s) [8] treatment without contact with HTC
Intervention [6] treatment by haemophilia treatment centre (HTC)
private physicians or haematologists= 13%,
hospital and nonhospital based clinics= 4%
hospitals or emergency rooms only= 8%
Sample size [9] 971
Sample size [7] 1979
Patient characteristics [10]
males with haemophilia A or B (not acquired)
Length of follow-up [11] 3 years
Outcome(s) measured [12] mortality
INTERNAL VALIDITY
Allocation [13] Non
randomised
Comparison of study groups
[14] Unclear
Blinding [15] none
Treatment/ measurement bias
[16] likely to be treated and
measured the same
Follow-up (ITT) [17]
Not stated
Overall quality assessment (descriptive) [18]
Selection bias unlikely
Confounding uncertain – adjusted for
Follow-up unclear
Patients representative
RESULTS
Outcome [19]
Mortality
Intervention group [20]
Control group [21]
149 (28.1%)
86 (38.3%)
Measure of
effect/effect size [22]
Benefits (NNT) [23]
RR= 0.7
95% CI [25]
95% CI= 0.6, 0.9
Harms (NNH) [24]
After multivariate
analysis
RR=0.6
95% CI [25]
95% CI= 0.5, 0.9
Clinical importance (1-4) [26] 1
Relevance (1-5) [27] 1
Any other adverse effects [28]
EXTERNAL VALIDITY
Generalisability [29] Good
Applicability [30] Good
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
221
STUDY DETAILS
Reference [1] (Soucie et al 2001)
Affiliation/source of funds [2] Hemophilia Surveillance System
Study design [3] Retrospective cohort study
Level of evidence [4] III-2
Location/setting [5] treatment centres in 6 states,
United States
Intervention [6] home therapy
Comparator(s) [8] No home therapy
Sample size [7] 1257
Sample size [9] 1393
Patient characteristics [10]
males with haemophilia A or B (not acquired) for whom home therapy status was able to be ascertained
median age= 19.9 years (range= 0.5 months to 95 years)
5.3% had inhibitors at baseline
80% haemophilia A
50% severe disease
Length of follow-up [11] 4 years
Outcome(s) measured [12] hospitalised bleeding complications
INTERNAL VALIDITY
Allocation [13] Non
randomised
Comparison of study groups
[14] those on home therapy
more likely to be white,
employed, have commercial
or government insurance,
higher levels of factor usage,
receiving HTC care, on
continuous prophylaxis and
inhibitors at baseline
Blinding [15] none
Treatment/ measurement bias
[16] likely to be treated and
measured the same
Follow-up (ITT) [17]
Good- (loss to followup only 3.3%)
Overall quality assessment (descriptive) [18] Good, 23/27
Selection bias unlikely
Confounding uncertain – adjusted for
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
No. of bleeding
complications
Intervention group [20]
Control group [21]
360 (28.6%)
448 (32.2%)
Measure of
effect/effect size [22]
Benefits (NNT) [23]
RR= 0.8
95% CI [25]
95% CI [25]
Harms (NNH) [24]
0.7, 0.9
p≤0.05
Clinical importance (1-4) [26] 1
95% CI [25]
Relevance (1-5) [27] 1
Any other adverse effects [28]
EXTERNAL VALIDITY
Generalisability [29] Good
Applicability [30] Good
Comments [31]
222
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Soucie et al 2004)
Affiliation/source of funds [2] Not stated
Study design [3] Retrospective cohort study
Intervention [6]
No factor concentrates
rFVIII
Location/setting [5] 130 haemophilia
treatment centres, United States
rFVIII + pdFVIII
pdFVIII
Sample size [7]
77
484
178
59
Patient characteristics
[10]
60 mild haemophilia, 13
moderate haemophilia, 4
severe haemophilia
61 mild haemophilia, 124
moderate haemophilia,
299 severe haemophilia
10 mild haemophilia, 28
moderate haemophilia,
140 severe haemophilia
15 mild haemophilia, 10
moderate haemophilia, 34
severe haemophilia
No inhibitors
31 with inhibitors
49 with inhibitors
11 with inhibitors
males with haemophilia,
born on or after February
1, 1993
Level of evidence [4] III-2
Length of follow-up [11] 2-7 years
Outcome(s) measured [12] B19 antibodies
INTERNAL VALIDITY
Allocation [13] unclear
Comparison of study groups
[14] No. sig diffs
Blinding [15] none
Treatment/ measurement bias
[16] possible
Follow-up (ITT) [17]
Adequate
Overall quality assessment (descriptive) [18]
Selection bias unlikely
Confounding uncertain
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
rFVIII alone
rFVIII + pdFVIII
pdFVIII
Odds ratio compared with
no exposure groups
0.8
1.9
7.6
(0.4-1.5)
(1.0-3.6)
(3.6-15.9)
p=0.5
p=0.05
p<0.001
(95% CI)
Benefits (NNT) [23]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26] 1
Relevance (1-5) [27]2
Any other adverse effects [28] B19 virus associated with loss of range of movement
EXTERNAL VALIDITY
Generalisability [29] Good
Applicability [30] Good
Comments [31] Shows clearly rFVIII is safer than pdFVIII
Use of recombinant and plasma-derived Factor VIII and IX
223
STUDY DETAILS
Reference [1] (Stajcic et al 1989)
Affiliation/source of funds [2] Not stated
Study design [3] Randomised controlled trial
Level of evidence [4] II
Location/setting [5] Out-patient hospital,
Yugoslavia
Intervention [6] Primary wound closure: EACA+ silk suture +
FVIII
Comparator(s) [8] Open wound group: EACA + FVIII
Sample size [7] 25
Sample size [9] 25
12 had one side each done both ways
Patient characteristics [10]
Intervention group – primary wound closure
15 severe, 2 moderate, 8 mild haemophilia
Comparator group(s) – Open wound
11 severe, 10 moderate, 4 mild haemophilia
Both at once9 severe, 1 moderate, 2 mild haemophilia
Length of follow-up [11] 5-7 days
Outcome(s) measured [12] Clot size, post operative bleeding, amount FVIII
required post-operatively
INTERNAL VALIDITY
Allocation [13]
randomised
Comparison of study groups
[14] No sig. Differences at
inception
Blinding [15] No
Treatment/ measurement bias
[16] Possible
Follow-up (ITT) [17]
Complete
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding uncertain
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Clot size
significantly
smaller in closed
wound group,
less bleeding.
Intervention group [20]
Control group [21]
Clot size large=15/25
Post op. bleeding=2/25
Post op FVIII=2.7 IU/kg
Benefits (NNT) [23]
Clot size large=1/22
Measure of effect/effect
size [22]
Post op
bleeding=0/25
Size of clot= chi2 test for
difference: p<0.001
95% CI [25]
Post op FVIII=0
Post op. bleeding=Fisher
exact test: p=0.72
95% CI [25]
Clinical importance (1-4) [26]
Harms (NNH) [24]
95% CI [25]
Relevance (1-5) [27]
Any other adverse effects [28]
EXTERNAL VALIDITY
Generalisability [29] Predominantly severe haemophilia. Not much information on participants so hard to assess external validity
Applicability [30] While using unvalidated measure, it does find clear patterns of benefit from primary wound closure
Comments [31]
224
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Tarantino et al 2004)
Affiliation/source of funds [2] several authors employed by Baxter Healthcare-product manufacturer,
rAHF-PFM Clinical study group
Study design [3] Randomised crossover trial
Level of evidence [4] II
Location/setting [5] 2 treatment centresAustria and Switzerland
Intervention [6] rAHF-PFM (no human or animal plasma)
Comparator(s) [8] rFVIII (Recombinate rAHF)
Sample size [7] 56-Part 1, 55 Part 3
Sample size [9] 56 Part 1, 55 Part 3
Patient characteristics [10]
PTPs≥ 150 ED, moderate-severe haemophilia
Exclude hypersensitivity to Recombinate rAHF, inhibitors >1.0 BU
Intervention group –
Comparator group(s) – same participants
Length of follow-up [11] 0.25-48 hours
Outcome(s) measured [12] PK-potency aPTT assay
INTERNAL VALIDITY
Allocation [13]
randomised
Comparison of study groups
[14] same participants
Blinding [15]
double-blinded
Treatment/ measurement bias
[16] same participants
Follow-up (ITT) [17]
30/55 (54.5%)
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding avoided
Follow-up adequate
Patients representative
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Recovery (IU/kg)
2.4±0.5
2.6±0.5
Measure of effect/effect
size [22]
Benefits (NNT) [23]
Not significant
95% CI [25]
95% CI [25]
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27]
Any other adverse effects [28] 19 mild-moderate adverse events related to AHF-PFM:
Taste perversion, headache, fever, diarrhoea, dizziness, hot flushes, pain (upper abdomen, lower chest), shortness of breath, sweating, nausea,
itching
EXTERNAL VALIDITY
Generalisability [29] see exclusions
Applicability [30]
Comments [31]
Use of recombinant and plasma-derived Factor VIII and IX
225
STUDY DETAILS
Reference [1] (White et al 1998)
Affiliation/source of funds [2]
The Recombinate PTP Study Group
Study design [3] Non-randomised crossover study
Level of evidence [4] III-2
Location/setting [5] Multicentre
Intervention [6] Phase 2 Trial
Comparator(s) [8]
50 IU/kg rFVIII (Recombinate)
50 IU/kg pdFVIII (Hemofil M)- solvent detergent treated
Sample size [7] 69- 67 severe, 2 moderate
Sample size [9]
Patient characteristics [10]
Severe-moderate haemophilia A, without inhibitors, PTPs
•
prophylaxis for surgery- Case series 13 participants
Intervention group – same group
Comparator group(s) –
Length of follow-up [11] 1) 24 hours, 2) prophylaxis 18 months,
30 months
Outcome(s) measured [12] ½ life, FVIII recovery, patient rated response,
blood loss (surgery)
INTERNAL VALIDITY
Allocation [13] Nonrandomised
Comparison of study groups
[14]
Blinding [15] No
Treatment/ measurement bias
[16]
Follow-up (ITT) [17]
Yes
Overall quality assessment (descriptive) [18]
Selection bias minimised
Confounding avoided
Follow-up adequate
External validity uncertain
RESULTS
Outcome [19]
Intervention group [20]
Control group [21]
Minimal blood loss
(surgery), as expected (0200 ml)
2.4± 0.97%
2.47± 0.33%
Measure of effect/effect
size [22]
2.42± 0.85
Benefits (NNT) [23]
95% CI [25]
95% CI [25]
p=0.59
Harms (NNH) [24]
95% CI [25]
Clinical importance (1-4) [26]
Relevance (1-5) [27]
Any other adverse effects [28] No. of adverse events= 13, (number of infusions= 13, 591)
Slight flushing, nausea, epistaxis, brief dizziness
EXTERNAL VALIDITY
Generalisability [29]
Applicability [30]
Comments [31]
226
Use of recombinant and plasma-derived Factor VIII and IX
STUDY DETAILS
Reference [1] (Zanon et al 2000)
Affiliation/source of funds [2] Not stated
Study design [3] Prospective cohort study
Level of evidence [4] III-2
Location/setting [5]
1 treatment centre, Italy
Intervention [6] For haemophilia
Comparator(s) [8] For non-haemophilia patients
FVIII or FIX to 30% of normal. 20 mg/kg tranexamic acid,
anaesthesia, fibrin sponge, silk suture
Silk sutures
Sample size [9] 184
Sample size [7] 77
Patient characteristics [10]
Intervention group – 32 severe haemophilia A, 39 moderate haemophilia A, 3 severe haemophilia B, 3 moderate haemophilia B
Comparator group(s) –184 subjects with bleeding disorders, liver disease or use of anticoagulant drugs
Length of follow-up [11] 7 days- 5 years
Outcome(s) measured [12] No. of bleeding complications
INTERNAL VALIDITY
Allocation [13]- non
random
Comparison of study groups
[14]
Blinding [15] No
Treatment/ measurement bias
[16] Unclear
Follow-up (ITT) [17]
Yes- complete
Overall quality assessment (descriptive) [18]
Selection bias uncertain
Confounding uncertain
Follow-up adequate
Patients representative
RESULTS
Outcome [19] Number of
bleeding complications
not statistically different
between groups
Intervention group [20]
2 bleeding
complications
Control group [21] 1
bleeding
complication
Measure of effect/effect
size [22] OR=0.2
Benefits (NNT) [23]
P=0.2
95% CI [25]
95% CI [25]
0.01-2.22
Clinical importance (1-4) [26]
Harms (NNH) [24]
95% CI [25]
Relevance (1-5) [27]
Any other adverse effects [28] 2 bleeding complications- late bleeding post extraction, and haematoma at site of anaesthesia injection
EXTERNAL VALIDITY
Generalisability [29] only moderate-severe haemophilia
Applicability [30]
Comments [31] More a dental surgery protocol than focusing on individual treatments
Use of recombinant and plasma-derived Factor VIII and IX
227
Explanatory Notes
STUDY DETAILS
[1] Full reference citation details
[2] Details of how the study was funded or other relevant affiliations of the authors
(designed to expose potential conflicts of interest, such as drug company funding for the
drug being trialled)
[3] The study type (eg RCT, case-control study, cohort study), with additional detail
where relevant
[4] As per the NHMRC levels of evidence, provided at pg-8 of the NHMRC Toolkit
publication: How to use the evidence: assessment and application of scientific evidence
[5] Country/setting (eg hospital, primary care, hospice)
[6] Provide detail on the intervention. This will generally be a therapeutic procedure such
as treatment with a pharmaceutical agent, surgery, a dietary supplement, a dietary change
or psychotherapy. Some other interventions are less obviously categorised as
interventions, such as early detection (screening) and patient educational materials. The
key characteristic is that a person or their environment is manipulated in the hope of
benefiting that person or reducing harm
[7] Number of participants enrolled in the intervention/treatment group
[8] The intervention (eg drug, therapy, placebo) used as a comparison in the study. There
may be more than one comparator
[9] Number of participants enrolled in the comparison/control group(s)
[10] Any factors that may confound/influence the results and/or the external validity
(see below) of the results (eg age, sex, comorbidities, obesity, existing medications,
previous surgery)
[11] Length of follow-up of the participants
[12] The outcomes studied (list all outcomes in terms of primary and secondary
outcomes). Indicate which outcomes are relevant to the review/guidelines inclusion
criteria
INTERNAL VALIDITY (QUALITY ASSESSMENT)
[13] The method used to assign patients to treatment or control groups (eg coin toss,
random number table, computer-generated random numbers, sealed envelopes). Also
indicate whether the allocation list was concealed (eg computerised random number
generation, administered from a central trial office, assigned locally)
[14] The results of the group analysis, noting any clinically or statistically significant
differences between the groups at study inception
[15] Whether the participants, outcome assessors and (if different) investigators were
blinded to the group allocation
[16] Indicate whether, aside from the experimental treatment, the groups were treated
and measured the same
[17] The proportion of participants that were followed up and whether all participants
were analysed according to the group to which they were initially allocated, regardless of
whether or not they dropped out, fully complied with the treatment, or crossed over and
received the other treatment (‘intention to treat analysis’ – ITT)
[18] Describe your assessment (in words) of the overall quality of the study. Is the study
quality good enough that you have confidence in the results?
228
Use of recombinant and plasma-derived Factor VIII and IX
RESULTS
[19] The outcome relevant for this entry in the database (Note: more than one table may
be required if there are several outcomes relevant to different clinical
questions/guidelines)
[20] For binary outcomes, show numbers of patients with the outcome. For continuous
outcomes, show means ± standard deviations; or medians and interquartile ranges
[21] For binary outcomes, show numbers of patients with the outcome. For continuous
outcomes, show means ± standard deviations; or medians and interquartile ranges. Add
number of columns as needed (eg 3-arm trials)
[22] Absolute and relative measures of effect eg risk differences (absolute risk reduction
or absolute risk increase), mean differences, relative risk, odds ratio
[23] A measure of benefit, when the treatment increases the probability of a good event.
The number needed to treat to benefit (NNT) = the number of participants who must
receive the treatment to create one additional improved outcome in comparison with the
control treatment; calculated as 1/absolute benefit increase, rounded up to the next
highest whole number
[24] A measure of harm, when the treatment increases the risk of specified adverse
outcomes of a condition or reduces the probability of a good event. The number needed
to treat to harm (NNH) = the number of patients who, if they receive the treatment,
would lead to one additional person being harmed compared with patients who receive
the control treatment; calculated as 1/absolute risk increase, rounded up to the next
highest whole number
[25] 95% confidence interval (CI) for all measures, if available, otherwise use P-value
[26] Insert the words corresponding to the appropriate rating from the scale provided at
pg-23 of the NHMRC Toolkit publication: How to use the evidence: assessment and application
of scientific evidence
[27] Insert the words corresponding to the appropriate rating from the scale provided at
pg-28 of the NHMRC Toolkit publication: How to use the evidence: assessment and application
of scientific evidence
[28] Information on any adverse events mentioned in the study
EXTERNAL VALIDITY
[29] Are the patients in the study so different from those being considered for the
guideline that the results may not be applicable to them?
[30] Will the potential benefits outweigh any potential harms of treatment in the
guideline population?
[31] Add your overall comments regarding the interpretation or implications of this study
Use of recombinant and plasma-derived Factor VIII and IX
229
Study profiles of included studies
Level of
evidence
Quality
score
Study Design
Study
Setting
Population
Treatment
Comparator
Outcome(s) assessed
Length of
follow-up
III-3
5/27
Historically
controlled
study
(Arkin et al. 1991)
Home-care
setting, worldwide
centres
14 non-bleeding PTPs with
moderate-severe haemophilia A,
aged 4–72, without inhibitors
rFVIII
(20–40 IU/kg, 3 x
week)
pdFVIII
Incremental FVIII
recovery (IU/kg-1) %
change
Not
reported
Adverse events
Historical controls: no description
provided
IV
IV
IV
3/3
2/3
2/3
Prospective
case series
(Auerswald et al.
2002)
Multicentre,
Germany, Austria
14 patients with vWD
Bolus doses of
43.5 IU FVIII/kg
and continuous
infusion of 3.4-9.6
IU FVIII/kg pdFVIII
N/A
55 hours
Adverse events
Prospective
case series
(Avanoglu et al.
1999)
1 treatment
centre, Turkey
22 patients: 21 haemophilia A, 1
haemophilia B, aged 6–17 years
Fibrin glue +
pdFVIII or pdFIX +
tranexamic mouth
wash
FVIII alone
Uncontrolled
before-andafter study
(Aygoren-Pursun
et al. 1997)
Multicentre,
Germany
39 PTPs with mild–severe
haemophilia A, aged 2–62 years,
treated for bleeding episodes,
prophylaxis and surgery
rFVIII (50 IU/kg,
Kogenate®)
N/A
Case series
Clinician judged
haemostasis, bleeding
complications
Bleeding episodes
12 days
Adverse events
Incremental FVIII
recovery (IU/kg-1)
12
months
Number of infusions per
bleeding episode
Adverse events
II
21/27
Randomised
single blinded
controlled trial
(Berntorp 1997)
Multicentre,
Sweden and the
Netherlands
12 PTPs with severe haemophilia
A, aged 17–64 years, without
inhibitors
rFVIII SQb
(50 IU/kg)
pdFVIII
(50 IU/kg,
Octonativ)
Incremental FVIII
recovery (IU/kg-1) %
change
50 hours
IV
2/3
Uncontrolled
before-andafter study
(Bjorkman et al.
2001)
Not stated
56 PTPs with moderate–severe
haemophilia B, aged 4–56 years,
without inhibitors
rFIX
N/A
Incremental FIX
recovery, % IU/kg-1
72 hours
IV
2/3
Prospective
case series
(Bray et al. 1994)
Multicentre,
United States
73 PUPs with moderate–severe
haemophilia A, aged 2 days–50
months, without inhibitors
rFVIII
N/A
Development of
inhibitors, no. of
exposure days prior to
inhibitor development
3-22
months
Adverse events
230
Use of recombinant and plasma-derived Factor VIII and IX
II
20/27
Randomised
crossover trial
(Cattaneo et al.
1989)
I treatment centre,
Milan, Italy
10 PTPs with severe vWD, (type
not specified) with prolonged
bleeding time (>30 minutes)
Cryoprecipitate +
DDAVP (0.3
µg/kg)
Cryoprecipitate +
saline
BT, vWF:Ag,
vWF:RiCof
Not stated
Crossover at
day 15
IV
1/3
Retrospective
case series
(Chediak et al.
1986)
1 treatment
centre, Chicago,
United States
6 women, 8 deliveries, 11
pregnancies
Whole blood, fresh
plasma,
cryoprecipitate,
curettage,
desmopressin
postpartum
N/A
Adverse events
Not stated
IV
2/3
Prospective
case series
(Chuansumrit et
al. 1993)
1 hospital,
Bangkok, Thailand
22 patients: 13 haemophilia A, 1
type 1 vWD, 8 with inherited or
acquired platelet dysfunction, aged
2–26 years
DDAVP (0.3–0.4
µg/kg) intravenous
N/A
Adverse events
48 hours
IV
3/3
Prospective
case series
(Collins et al.
2004)
Multicentre,
United Kingdom
18 acquired haemophilia: 14 no
underlying disorder, 1 gastric
carcinoma, 1 polymyalgia
rheumatica, 1 Castlemanns
disease, 1 Depoxil, aged 38–87,
median 70 years
Prednisolone +
cyclophosphamide
N/A
Adverse events
12
months
IV
1/3
Retrospective
case series
(Daffos et al.
1988)
1 treatment
centre, Paris,
France
93 pregnant women with
haemorrhagic disorders:
Haemophilia A:
35 obligate and 44 putative
carriers, Haemophilia B:
8 women obligate and 4 putative
carriers, vWD:
1 x Type 1; 1 x Type 2A
Antenatal
diagnosis: fetal
blood sampling
N/A
Effectiveness of
antenatal diagnosis,
Adverse events
Not stated
68 patients: 32 mild and 8
moderate–severe haemophilia A,
13 type 1 vWD, 7 type 2A vWD, 1
type 2B vWD, aged 2–66 years
DDAVP (0.3
µg/kg) intravenous
N/A
Clinician judged
haemostasis, need for
alternative therapies
7 days
26 patients, 21 mild haemophilia A,
5 moderate haemophilia A
Intravenous
DDAVP (0.3
µg/kg)
IV
III-2
2/3
17/27
Prospective
case series
Non
randomised
controlled trial
(de la Fuente et
al. 1985)
(De Sio et al.
1985)
2 treatment
centres, United
States
1 treatment
centre, Rome,
Italy
Use of recombinant and plasma-derived Factor VIII and IX
Treatment as
required:
cryoprecipitate
Adverse events
Subcutaneous
DDAVP (0.3
µg/kg)
FVIII:C levels, post/pre
FVIII:C ratio
2 hours
Side effects
231
IV
4/6
Systematic
review
(Delgado et al.
2003)
Mixed
20 retrospective or prospective
case series of 249 patients with
acquired haemophilia, median age
64 years (8–93 years)
DDAVP, human
FVIII, porcine FVIII,
aPCCs, rFVIIa,
intravenous
immunoglobulin,
plasmapheresis and
immunoadsorption,
FVIII immune
tolerance
N/A
Response rates to
treatment of bleeding
episodes and to
eradication therapy
Not stated
IV
2/3
Prospective
case series
(Dewald et al.
1980)
1 clinical research
centre, Ohio,
United States
5 patients: 4 haemophilia A, 1
vWD
DDAVP (0.4 µg/kg)
intranasal
N/A
Side effects
24 hours
IV
2/3
Prospective
case series
(Djulbegovic et al.
1996)
Unclear—1
treatment centre?
United States
8 patients with haemophilia B
EACA or
tranexamic acid ±
FIX
N/A
Adverse events
30 days
IV
2/3
Prospective
case series
(Dobrkovska et al.
1998)
1 treatment
centre, Chicago,
United States
6 patients with vWD: 2 x Type 1B,
2 x Type 2A, 2 x Type 3
80 IU vWF:RcoF kg1 and 32 IU FVIII:C
kg-1 pdFVIII
N/A
In vivo recovery and
half lives of
vWF:Ag, vWF:Rcof
and FVIII:C, BT
60 hours
IV
2/3
Retrospective
case series
(Dobrkovska et al.
1998)
18 treatment
centres, Canada
97 patients with vWD: 32 x Type 1,
5 x Type 2A, 18 x Type 2B, 28 x
Type 3,
14 x Other
11.9–222.8 IU
vWF:Rcof/kg
pdFVIII for surgery,
prophylaxis or
bleeding episodes
N/A
Clinician judged
haemostasis, Median in
vivo recovery FVIII:C,
vWF:RcoF
IV
2/3
Retrospective
case series
(Dunn et al. 2000)
1 treatment
centre,
Washington DC,
United States
40 patients: 24 types 1 and 2 vWD,
16 mild–moderate haemophilia A
and symptomatic haemophilia A
carriers aged 5–58 years
DDAVP
intranasally, 150 µg
if under 50 kg, 300
µg if over 50 kg
N/A
Adverse events
Not stated
II
22/27
Randomised
crossover trial
(Ewenstein et al.
2002)
15 treatment
centres, United
States
38 non-bleeding PTPs with severe
haemophilia B, aged 7–75 years,
without inhibitors
rFIX
pdFIX
Incremental FIX
recovery, % IU/kg-1 %
change
48 hours
IV
2/3
Retrospective
case series
(Federici et al.
2000)
1 haemophilia
centre, Milan, Italy
63 patients with vWD: 31 x Type 1,
22 x Type 2, 10 x Type 3
Tranexamic acid±
fibrin glue,± DDAVP
or pdFVIII
N/A
Number of bleeding
complications
8 days
Adverse events
232
21 days
Adverse events
Use of recombinant and plasma-derived Factor VIII and IX
IV
1/3
Retrospective
case series
(Federici et al.
2002)
Multicentre, Italy
22 patients with vWD: 9 x Type 1,
7 x Type 2B, 6 x Type 3
17–92 IU
FVIII:C/kg/day
pdFVIII
N/A
Clinician judged
haemostasis
26 days
No. of bleeding
episodes controlled with
1 infusion
Adverse events
IV
3/3
Before-andafter study
(Federici et al.
2004)
5 treatment
centres, Europe
66 patients with type severe type 1
or 2 vWD, BT more than 15
minutes, aged 12–65 years
DDAVP (0.3 µg/kg)
intravenous infusion
N/A
No. responsive to
DDAVP
4 hours
Responsive= 3-fold
increase post/pre
treatment in FVIII:C and
vWF:Rco + BT ≤12
minutes
II
21/27
Randomised
single blinded
controlled trial
(Fijnvandraat et
al. 1997)
Multicentre,
Sweden and the
Netherlands
12 PTPs with severe haemophilia
A, aged 17–64 years, without
inhibitors
rFVIII SQb
(50 IU/kg)
pdFVIII
(50 IU/kg,
Octonativ)
Incremental FVIII
recovery (IU/kg-1) %
change
50 hours
IV
1/3
Retrospective
case series
(Franchini et al.
2003)
3 treatment
centres, Italy
26 patients with vWD: 19 x Type 1,
7 x Type 2B
125.0-976.4 IU
vWF:Rcof/kg
pdFVIII for major
surgery, 42.9–173.3
IU vWF:Rcof/kg
pdFVIII for minor
surgery, 27.2-160.0
IU vWF:Rcof/kg
pdFVIII for dental
extractions
N/A
vWF:Ag, vWF:Ag,
FVIII:C levels pre/post,
in vivo recovery of
vWF:Ag, amount of
pdFVIII used
23 days
Adverse events
IV
2/3
Prospective
case series
(Ghirardini et al.
1987)
Unclear
25 patients: 23 mild haemophilia A,
2 moderate haemophilia A
DDAVP (0.3 µg/kg)
subcutaneous
N/A
Adverse events
60
minutes
IV
2/3
Prospective
case series
(Ghirardini et al.
1988)
Unclear
24 patients: 16 mild haemophilia A,
2 moderate haemophilia A, 6 type
1 vWD patients
DDAVP
subcutaneous (0.3
µg/kg) + tranexamic
acid + fibrin glue for
dental
N/A
No. of bleeding
complications
7 days
Use of recombinant and plasma-derived Factor VIII and IX
Adverse events
233
IV
1/3
Retrospective
case series
(Giangrande
2002)
19 treatment
centres, Europe
and Israel
31 PUPs and MTPs with severe
haemophilia A, aged ≤ 4 years,
without inhibitors
rFVIII
N/A
Number of infusions per
bleeding episode
2 years
Development of
inhibitors
No. of exposure days
prior to inhibitors
III-2
15/27
Non
randomised
controlled trial
(Giles et al. 1998)
Multicentre,
Canada
872 PTPs with mild–severe
haemophilia
rFVIII
Affinity purifiedFVIII
Inhibitor development
associated with factor
replacement therapy
2 years
IV
2/3
Prospective
case series
(Gill et al. 2002)
1 treatment
centre,
25 patients: 9 haemophilia A, 16
vWD
DDAVP intranasal
(150 µg for under
50 kg)
N/A
Adverse events
24 hours
IV
3/3
Prospective
case series
(Gill et al. 2003)
19 treatment
centres, United
States
33 patients with vWD: 9 x Type 1,
4 x Type 2A, 7 x Type 2B, 27 x
Type 3, 6 x Other
Loading dose 25.7–
143.2 IU/kg plus
16/4–182.9 UK/kg
maintenance dose
pdFVIII
N/A
Clinician judged
haemostasis
>7 days
Adverse events
IV
2/3
Retrospective
case series
(Greer et al. 1991)
1 treatment
centre, Glasgow,
United Kingdom
18 obligate carriers of haemophilia
A, 34 pregnancies, 5 obligate
carriers of haemophilia B, 11
pregnancies
Treatment as
required:
cryoprecipitate,
tranexamic acid,
fresh frozen plasma,
whole blood
transfusions
N/A
Adverse events
30 year
period
IV
3/3
Prospective
case series
(Gringeri et al.
2004)
Multicentre, Italy
25 PTPs with severe haemophilia
A, aged 6–60 years, without
inhibitors
rFVIII
N/A
Number of infusions per
bleeding episode
6 months
Inhibitor development
No. of exposure days
prior to inhibitors
IV
IV
234
2/3
3/6
Retrospective
case series
(Gringeri et al.
2004)
Multicentre, Italy
Systematic
review
(Gruppo et al.
2003)
Mixed
94 PTPs with mild–severe
haemophilia A, without inhibitors,
treated for bleeding episodes or
prophylaxis
rFVIII
13 observational studies
B-domain deleted
rFVIII for prophylaxis
N/A
Inhibitor development
2 years
No. of exposure days
prior to inhibitors
Full length FVIII
for prophylaxis
(pdFVIII or
rFVIII)
No. of breakthrough
bleeding episodes, half
life of FVIII
12 year
period
Use of recombinant and plasma-derived Factor VIII and IX
IV
IV
3/3
2/3
Prospective
case series
Retrospective
case series
(Hoyer et al.
1985)
(Kadir et al. 1997)
Multicentre,
United States and
Canada
92 pregnancies at risk of
haemophilia A
1 treatment
centre, London,
United Kingdom
24 haemophilia A and 8
haemophilia B obligate carriers, 82
pregnancies
Antenatal diagnosis
N/A
Treatment as
required
Antenatal diagnosis
Effectiveness of
antenatal diagnosis
6 year
period
Adverse events
N/A
Treatment as
required:
Uptake and results of
antenatal diagnosis
10 year
period
Adverse events
DDAVP, vWF rich
concentrates, rFVIII,
oral contraceptive
pill, fresh frozen
plasma
IV
II
2/3
24/37
Retrospective
case series
Randomised
controlled trial
(Kadir et al. 1998)
(Kadir et al. 2002)
1 treatment
centre, London,
United Kingdom
31 women with vWD (27 Type 1; 2
Type 2; 2 Type 3), 11 with XI
deficiency, 84 pregnancies, 85
foetuses
Treatment as
required:
Home treatment,
United Kingdom
39 women with menorrhagia. Mildmoderate vWD, heterozygote FXIdeficiency, and carriers of
haemophilia
DDAVP (300 µg)
intranasal
N/A
Mode of delivery
Adverse events
17 year
period
DDAVP, vWF rich
concentrate, fresh
frozen plasma,
tranexamic acid, oral
contraceptive pill,
blood transfusions
Placebo nasal
spray
Mean difference in
Pictorial Blood
Assessment Chart
scores
2 months
Side effects
III-1
21/27
Quasirandomised
single-blinded
(investigator)
controlled trial
(Kelly et al. 1997)
1 treatment
centre,
Philadelphia,
United States
10 non-bleeding PTPs with severe
haemophilia A, aged 6–12, without
inhibitors
rFVIII
(50 IU/kg,
RecombinateTM)
pdFVIII
(50 IU/kg,
Hemofil®M)
Incremental FVIII
recovery (IU/kg-1)%
change
72 hours
II
21/27
Randomised
double-blinded
crossover trial
(Kisker et al.
2003)
1 treatment
centre, Iowa City,
United States
15 non-bleeding severe
haemophilia B patients, aged >12
years, without inhibitors
rFIX
(50 IU/kg,
BeneFIX®)
pdFIX
(50 IU/kg,
Mononine®)
Incremental factor
recovery, IU/kg-1 , %
change
7-30 days
III-3
11/27
Historical
control study
(Kreuz et al. 2002)
1 treatment
centre, Frankfurt,
Germany
72 PUPs with moderate–severe
haemophilia A, without inhibitors,
treated for bleeding episodes,
prophylaxis, or surgery
rFVIII
pdFVIII
Development of
inhibitors
23 year
period
Use of recombinant and plasma-derived Factor VIII and IX
No. of exposure days
prior to inhibitors
235
IV
1/3
Before and
after study
(Leissinger et al.
2001)
Multicentre,
United States
Prospective
case series
IV
IV
IV
2/3
2/3
6/6
Retrospective
case series
Retrospective
case series
Systematic
review
(Lillicrap et al.
2002)
(Ljung et al. 1994)
(Lloyd Jones et al.
2003)
18 treatment
centres, Canada
Multicentre,
Sweden
Mixed
333 patients enrolled. 124 mild
haemophilia A, 135 mild type 1
vWD, 37 moderate type 1 vWD, 23
haemophilia carriers, 14 other
bleeding disorders, aged 5–64
years
DDAVP (150 µg)
intranasal
97 patients with vWD: 32 x Type 1,
5 x Type 2A, 18 x Type 2B, 28 x
Type 3,
14 x Other
11.9–222.8 IU
vWF:Rcof/kg dFVII
for surgery,
prophylaxis or
bleeding episodes
N/A
117 pregnant women, 117
deliveries of haemophilic children
Blood sampling,
vitamin K injections
N/A
Adverse events
20 year
period
101 haemophilia A (77 severe, 24
moderate)
Treatment as
required:
16 haemophilia B (12 severe, 4
moderate)
Whole blood
transfusion
52 relevant papers: 8 randomised
controlled trials, 2 non randomised
controlled trials, 1 survey, 39 case
series/uncontrolled prospective
studies
Human FVIII,
porcine FVIII, PCCs,
aPCCs, rFVIIa,
plasmapheresis,
extracorporeal
protein A adsorption,
cimetidine or
immunosuppression
N/A
No. patients with
haemostatic control
Not stated
N/A
Clinical judged
haemostasis
48 hours
Adverse events
Clinician judged
haemostasis, Median in
vivo recovery FVIII:C,
vWF:RcoF
21 days
Adverse events
Adverse events
IV
2/3
Retrospective
case series
(Lorenzo et al.
2001)
2 treatment
centres, Spain
62 consecutive severe
haemophilia A patients
FVIII prophylaxis
N/A
Cumulative incidence of
inhibitors at 3 years
from first FVIII exposure
≤75 days
IV
2/3
Prospective
case series
(Lusher et al.
1993)
Multicentre,
United States,
Canada, Europe,
United Kingdom
95 PUPs with mild-severe
haemophilia A, aged 0–658
months, treated for bleeding
episodes or prophylaxis
rFVIII
N/A
Development of
inhibitors
3.5 years
236
Adverse events
Use of recombinant and plasma-derived Factor VIII and IX
IV
2/3
Prospective
case series
(Lusher et al.
2004)
28 treatment
centres, Europe
and United States
102 PUPs with mild–severe
haemophilia A, without inhibitors,
treated for bleeding episodes,
prophylaxis or surgery
rFVIII
N/A
Number of infusions per
bleeding episode
Development of
inhibitors
2–5 years
or 100
exposure
days
No. of exposure days
prior to inhibitors
Adverse events
II
23/27
Randomised
crossover trial
(Mannucci et al.
1987)
Multicentre,
Europe
14 mild and moderate haemophilia
A (median FVIII:C level 7 U/dL,
range 2–31)
Intravenous DDAVP
(0.3 µg/kg)
Subcutaneous
DDAVP (0.3
µg/kg)
Peak FVIII:C, half life of
FVIII:C
24 hours
IV
2/3
Before and
after study
(Mannucci et al.
1992)
1 treatment
centre, Milan, Italy
37 patients, 22 with mild
haemophilia A, 15 with mild type 1
vWD, aged 16–63 years
DDAVP (0.3 µg/kg)
intravenous
N/A
FVIII:C, vWF:Ag, RiCof,
BT post/pre treatment
ratios
4 days
Prospective
case series
III-3
16/27
Historically
controlled
study
(Mannucci et al.
1994)
13 treatment
centres in Europe
and United States
PTPs with moderate-severe
haemophilia A, 30 HIV
seropositive, 31 HIV seronegative
rFVIII
pdFVIII
Change in immune
status
Median
3.5 years
II
22/27
Randomised
crossover trial
(Mannucci et al.
2002)
Multicentre,
United States,
Europe
12 patients with type 3 vWD
60 vWF:Rcof IU/kg
pdFVIII-SD
60 vWF:Rcof
IU/kg pdFVIIISD/HT
Bleeding time, in vivo
half lives of FVIII:C,
vWF:Ag, vWF:Rcof
Crossover
7–186
days later
24 hours
IV
3/3
Before and
after study
(Mannucci et al.
2002)
Multicentre,
United States,
Europe
81 patients with vWD: 15 x Type 1,
29 x Type 2A, 5 x Type 2B, 32 x
Type 3
pdFVIII-SD
pdFVIII-SD/HT
Prospective
case series
IV
1/3
Before and
after study
1/3
Retrospective
case series
50 hours
Adverse events
(Mariana et al.
1984)
Multicentre, Bari
and Rome, Italy
Retrospective
case series
IV
Clinical judged
haemostasis, need for
alternative therapy, BT
(Mohri 2002)
?
Japan
Use of recombinant and plasma-derived Factor VIII and IX
43 patients, 21 mild haemophilia A,
2 moderate haemophilia A, 20
vWD (types 1 and 2)
DDAVP (0.3 µg/kg)
intravenous
3 patients with menorrhagia, 1 type
1 vWD, 2 type 2A vWD, aged 23–
43 years
Single dose
tranexamic acid (3 g
for 1–5 days)
DDAVP (0.4
µg/kg)
intravenous
Cessation/prevention of
bleeding
N/A
Control of menorrhagia
after treatment
11 days
Adverse events
5 years
Adverse events
237
III-2
16/27
Nonrandomised
crossover trial
(Morfini et al.
1992)
Multicentre,
Europe and
United States
47 PTPs with severe haemophilia
A, without inhibitors
rFVIII
(50 IU/kg,
RecombinateTM)
pdFVIII
(50 IU/kg,
Hemofil®)
Incremental FVIII
recovery (IU/kg-1) %
change, In vivo
recovery (%)
24 hours
IV
2/3
Retrospective
case series
(Morrison et al.
1993)
47 treatment
centres in Europe
and North
America
65 patients with acquired
haemophilia, associations:
unknown= 55%, pregnancy=11%,
malignancy=12%, connective
tissue or autoimmune
disease=17%, skin disease=2%,
drug treatment =3%
Porcine FVIII
N/A
Clinical judged
haemostasis
Not stated
IV
3/3
Before and
after study
(Nolan et al. 2000)
1 treatment
centre, Ireland
133 patients with bleeding
disorders. 91 type 1 vWD, 20 mild
haemophilia A, 22 platelet function
disorders
DDAVP (0.3 µg/kg)
continuous infusion
N/A
No. responsive to
DDAVP
Not stated
Prospective
case series
Response= increase of
FVIII:C into normal
range for haemophilia,
increase of vWF:Ag,
vWF:FVIIIB and FVIII:C
into normal range for
vWD. Normal range for
all = 0.5-1.5 IU/dl
90
minutes?
Side effects
IV
2/3
Retrospective
case series
(Nitu-Whalley et
al. 2001)
1 treatment
centre, London,
United Kingdom
27 patients with types 1 (93%) and
2 (7%) vWD most of whom had
responded to DDAVP, aged 14–57
years
Intravenous DDAVP
(0.3 µg/kg) +
tranexamic acid for
mucosal surgery
N/A
Clinician judged
haemostasis
16 days
IV
1/3
Retrospective
case series
(Ong et al. 1998)
1 hospital, Belfast,
United Kingdom
4 patients with menorrhagia, 1 type
1 vWD, 2 type 2A vWD, 1 type 2B
vWD, aged 17–42 years.
Single dose
tranexamic acid (4 g
for 3–5 days)
N/A
Control of menorrhagia
after treatment
4 years
22 patients with severe
haemophilia A and 3 children with
severe haemophilia B, aged 1.7–
20.4
Prophylaxis with
rFIX
N/A
IV
238
2/3
Uncontrolled
before-andafter study
(Panicker et al.
2003)
1 treatment
centre, Detroit,
United States
Adverse events
Incremental FIX
recovery, IU/kg-1
13 year
period
Use of recombinant and plasma-derived Factor VIII and IX
III-3
15/27
Historical
control study
(Poon et al. 2002)
16 treatment
centres, Canada
PTPs with mild–severe
haemophilia B, aged 1–74 years
rFIX
pdFIX
Incremental FIX
recovery, % IU/kg-1 %
change
IV
2/3
Prospective
case series
IV
2/3
Uncontrolled
before-andafter study
(Ragni et al. 2002)
12 treatment
centres, Europe
and United States
26 PTPs with mild–severe
haemophilia B and 2 female
haemophilia B carriers
rFIX for surgery
N/A
Adverse events
30 days
IV
2/3
Prospective
case series
(Rakocz et al.
1993)
1 treatment
centre, Israel
80 patients with bleeding
disorders, 37 severe haemophilia
A/B, 4 mild haemophilia A/B, 10
mild/moderate vWD, 29 other
Fibrin glue used
with aprotinin
concentration 1,000
or 10,000 KIU/ml ±
ranexamic acid
mouthwash
N/A
Adverse events
10 days
IV
2/3
Prospective
case series
(Rodeghiero et al.
1996)
17 treatment
centres, Italy
79/169 patients, 43 vWD, 36
haemophilia A
DDAVP (20 µg for
>70 kg or 40 µg for
<70 kg)
subcutaneous in
home treatment
N/A
Side effects
12
months
IV
2/3
Prospective
case series
(Rose & Aledort
1991)
1 treatment
centre? United
States
22 patients, 11 vWD, 8 mild
haemophilia A, 3 symptomatic
carriers of haemophilia A
DDAVP (300 µg for
adults, 150 µg for
children—
unspecified age)
intranasally
N/A
Adverse events
2–4 days
IV
2/3
Uncontrolled
before-andafter study
(Roth et al.
2001a)
20 treatment
centres, Europe
and United States
56 PTPs with moderate–severe
haemophilia B, aged 4–56 years,
without inhibitors
rFIX
N/A
Incremental FIX
recovery, % IU/kg-1
2 years
≤5 years
Adverse events
Case series
Number of infusions per
bleeding episode
Development of
inhibitors
No. of exposure days
prior to inhibitors
Adverse events
Use of recombinant and plasma-derived Factor VIII and IX
239
IV
2/3
Prospective
case series
(Rothschild et al.
1998)
21 treatment
centres, France
50 PUPs with severe haemophilia
A, aged 1–43
rFVIII
N/A
Development of
inhibitors
12
months
No. of exposure days
prior to inhibitors
IV
1/3
Retrospective
case series
(Sallah 2004)
3 treatment
centres, United
States
34 patients with acquired
haemophilia, associations: 10 x
none, 3 x post partum, 1 x drug
reaction, 6 x cancer, 3 x lupus, 10
x other
75 IU/kg of FEIBA
N/A
% with complete or
partial response (control
of bleeding)
48 hours
Side effects
IV
3/3
Prospective
case series
(Santagostino et
al. 2003)
1 treatment
centre, Milan, Italy
27 patients with severe
haemophilia who had difficulties in
venipuncture
Arteriovenous
fistulae as vascular
access
N/A
No. of bleeding
complications, failure of
permanent vascular
access, thrombotic
complications, shuntrelated complications
5–44
months
IV
2/3
Uncontrolled
before-andafter study
(Saulnier et al.
1994)
3 hospitals,
France
14 vWD patients, mean age 24
years
DDAVP (0.3 µg/kg)
+ tranexamic acid +
fibrin glue
N/A
Number without
bleeding complication,
Post/pre ratio of FVIII:C,
vWF:Ag, RiCof
15 days
IV
2/3
Uncontrolled
before-andafter study
(Scharrer 2002;
Scharrer et al.
2000)
5 treatment
centres, Europe
15 PTPs and 7 PUPs with severe
haemophilia A, without inhibitors
rFVIII
N/A
Adverse events
24
months
II
13/27
Randomised
double blind
controlled trial
(Schiavoni et al.
1983)
Haemophilia
Summer camp,
Sweden
26 patients, 17 severe haemophilia
A, 2 mild haemophilia A, 7 severe
haemophilia B
Tranexamic acid 25
mg/kg 3x daily as
prophylaxis
Placebo
Number of bleeding
episodes, amount of
FVIII used
2 weeks
IV
3/3
Before-andafter study
(Schulman et al.
1991)
1 treatment
centre, Stockholm,
Sweden
370 patients with disorders of
primary haemostasis. 133 vWD,
237 platelet function defects, aged
3–80 years
DDAVP (0.2–0.3
µg/kg) route of
administration not
specified
N/A
No. responsive to
DDAVP
24 hours
Case series
Responsive= return to
normal or reduction by
more than 20% of BT,
or greater than 2-fold
increase in vWF.
‘Normal’ not stated
Side effects
240
Use of recombinant and plasma-derived Factor VIII and IX
III-2
19/37
Nonrandomised
crossover trial
IV
3/3
Prospective
case series
III-2
19/37
Nonrandomised
crossover trial
IV
3/3
Prospective
case series
(Schwartz et al.
1990)
(Seremetis et al.
1999)
Multicentre,
Worldwide
Multicentre- home
treatment program
17 non-bleeding PTPs with
moderate-severe haemophilia A,
without inhibitors
rFVIII
(50 IU/kg)
58 PTPs with moderate–severe
haemophilia A, without inhibitors,
treated
rFVIII
(50 IU/kg)
17 non-bleeding PTPs with
moderate-severe haemophilia A,
without inhibitors
rFVIII
(50 IU/kg)
pdFVII
(50 IU/kg, Koate
HS)
Incremental FVIII
recovery (IU/kg-1) %
change, number of
infusions per bleeding
episode
48 hours
Development of
inhibitors
Adverse events
pdFVII
(50 IU/kg, Koate
HS)
58 PTPs with moderate–severe
haemophilia A, without inhibitors,
treated
Incremental FVIII
recovery (IU/kg-1) %
change, number of
infusions per bleeding
episode
3 years
Development of
inhibitors
Adverse events
IV
1.5/3
Retrospective/
Prospective
case series
(Sindet-Pedersen
et al. 1988)
1 hospital,
Denmark
15 patients: 13 haemophilia A, 2
haemophilia B
Tranexamic acid
orally (25 mg/kg 4x
daily) or as
mouthwash (10 ml
of 5% tranexamic
acid solution, 4x
daily for 2 minutes)
± FVIII
N/A
Adverse events
Not stated
III-2
21/27
Retrospective
cohort study
(Soucie et al.
2004)
130 treatment
centres, United
States
818 males with haemophilia A,
born after 1 February 1993
rFVIII, rFVIII +
pdFVIII, pdFVIII
No FVIII
B19 parvovirus
antibodies
2–7 years
III-2
22/27
Retrospective
cohort study
(Soucie et al.
2000)
Both haemophilia
and nonhaemophilia
treatment centres,
6 states in United
States
2950 males with haemophilia A or
B, identified over 3 year period
Haemophilia
treatment centre
Non-haemophilia
treatment centre
Mortality
3 years
III-2
23/27
Retrospective
cohort study
(Soucie et al.
2001)
Medical treatment
centres, 6 states
of United States
2650 males with haemophilia A or
B
Home treatment
No home
treatment
Haemorrhagic bleeding
complications
4 years
Use of recombinant and plasma-derived Factor VIII and IX
241
II
14/27
Randomised
controlled trial
(Stajcic et al.
1989)
1 treatment
centre, Yugoslavia
62 patients with mild–severe
haemophilia A, undergoing dental
extractions
Open wound:
EACA + FVIII
Closed wound:
EACA + silk
suture + FVIII
Clot size, post operative
bleeding, amount FVIII
required postoperatively
5–7 days
IV
2/3
Retrospective
case series
(Stasi et al. 2004).
1 treatment
centre,
10 acquired haemophilia: 6 with no
underlying disorder, 1 x
rheumatoid arthritis, 1 x prostate
carcinoma, 1 x low-grade nonHodgkin lymphoma, and 1 after
pregnancy, aged 27–78 years,
median 61 years
Rituximab
immunosuppressan
t 375 mg/m2
N/A
No. achieved partial or
complete remission of
inhibitors
12–41
months
Side effects
IV
1/3
Retrospective
case series
(Stieltjes et al.
2004)
9 treatment
centres, Europe
16 PTPs with mild–severe
haemophilia A, without inhibitors
Recombinant Bdomain deleted
FVIII
N/A
Adverse events
3 year
period
II
25/27
Double
blinded
randomised
controlled trial
(Tarantino et al.
2004)
2 treatment
centres, Austria
and Switzerland
56 PTPs with moderate–severe
haemophilia A, aged 10–65 years,
without inhibitors, with ≥150
exposure days
R-FVIII
(50 IU/kg)
rAHF-PFMa (50
IU/kg)
Incremental FVIII
recovery (IU/kg-1), %
change
0.25–48
hours
IV
3/3
Case series
Development of
inhibitors
Adverse events
IV
2/3
Prospective
case series
(Thompson et al.
2004)
28 treatment
centres, United
States
39 patients with vWD: 16 x Type 1,
4 x Type 2A, 5 x Type 2B, 8 x
Type 3, 6 x Other
32.5–216.8 IU
vWF:Rco/ kg
pdFVIII
N/A
No. and dose of
infusions needed,
clinician judged
haemostasis
>21 days
Adverse events
IV
6/6
Systematic
review
(Valentino et al.
2004)
Mixed
2074 patients with haemophilia A,
B von Willebrand or other
congenital coagulation disorders
from 48 studies
Central venous
access devices
(CVAD), fully
implanted
Central venous
access devices,
external
Infections, thrombotic
complications and other
adverse events
Mean 578
days per
CVAD
IV
2/3
Retrospective
case series
(van der Bom et
al. 2003)
1 treatment
centre, Utrecht,
The Netherlands
81 consecutive severe
haemophilia A patients
FVIII prophylaxis
N/A
Cumulative incidence of
inhibitors at 100 days
from first FVIII exposure
3–26
years
(mean=
16 years)
Mean number of
infusions before
antibodies
242
Use of recombinant and plasma-derived Factor VIII and IX
IV
2/3
Before-andafter study
(Warrier & Lusher
1983)
Prospective
case series
1 hospital,
Michigan, United
States
38 patients, 31 vWD, 7 mild–
moderate haemophilia A and 3
normal subjects
DDAVP via
intranasal drip (2–4
µg/kg) or
intravenous (0.2–
0.3 µg/kg)
N/A
% Rise in FVIII:C,
FVIII:RAg, FVIII:RCof,
BT
270
minutes
Side effects
24 hours
IV
2/3
Retrospective
case series
(Wiestner et al.
2002)
2 treatment
centres, United
States
4 patients with acquired
haemophilia, associations: 1 x
chronic renal failure, 1 x ascites
and lymphadenopathy, 1 x lupus, 1
x mild haemophilia
Rituximab
immunosuppressan
t 375 mg/m2
N/A
Adverse events
7–12
months
IV
2/3
Prospective
case series
(White et al. 1997)
Multicentre
69 PTPs with moderate–severe
haemophilia A, without inhibitors
rFVIII
N/A
Development of
inhibitors
24 hours–
30
months
Adverse events
II
17/27
Randomised
crossover trial
(White et al. 1998)
20 treatment
centres, Europe
and United States
11 PTPs with haemophilia B
rFIX
pdFIX
Incremental FIX
recovery, % IU/kg-1, %
change
72 hours
IV
2/3
Case series
(White et al. 1998)
20 treatment
centres, Europe
and United States
56 PTPs with moderate–severe
haemophilia B, aged 4–56 years,
without inhibitors
rFIX
N/A
Number of infusions per
bleeding episode
2 years
Development of
inhibitors
IV
6/6
Systematic
review
(Wight et al. 2003)
Mixed
1 semi-structured review, 29 case
series, 8 case reports, 4 registries
(probably overlap)
Any form of
immune tolerance
induction: Bonn,
Malmo, Low-dose,
Porcine FVIII
N/A
Success rates
(undetectable inhibitors)
≤103
months
IV
2/3
Prospective
case series
(Yoshioka et al.
2001)
Multicentre, Japan
20 PTPs with moderate–severe
haemophilia A, aged 12–55 years,
without inhibitors, treated for
bleeding episodes
rFVIII-FSd (10-50
IU/kg, Kogenate®)
N/A
Incremental FVIII
recovery (IU/kg-1)
12–24
weeks
Use of recombinant and plasma-derived Factor VIII and IX
Number of infusions per
bleeding episode
243
IV
2/3
Prospective
case series
(Yoshioka et al.
2003)
33 treatment
centres in Japan
43 PUPs and MTPs with mild–
severe haemophilia A, aged 3–386
months, treated with rFVIII
(Kogenate®) for bleeding episodes
rFVIII
N/A
Number of infusions per
bleeding episode
3
months–
Development of
inhibitors
2 years
No. of exposure days
prior to inhibitors
III-2
16/27
Cohort study
(Zanon et al.
2000)
1 treatment
centre, Padova,
Italy
71 haemophilia A and 6
haemophilia B patients undergoing
dental extractions
Controls: 184 non-haemophilic
patients undergoing dental
extractions
rFVIII /rFIX +
tranexamic acid +
fibrin sponge + silk
suture
Silk suture
No. of bleeding
complications
7 days
5 year
period
vWD= von Willebrand disease SD= solvent detergent; HT= heat treated, BT= bleeding time, FVIII:RAg= factor VIII antigen, FVIII:RCof= factor VIII ristocetin cofactor , FVIII:C= factor VIII coagulant activity, vWF= von Willebrand factor, FVIII=
factor VIII , vWD= , pdFVIII= plasma derived factor VIII , PTP= previously treated patients, N/A= not applicable
244
Use of recombinant and plasma-derived Factor VIII and IX